Next Article in Journal
Quasi-Static and Dynamic Measurement Capabilities Provided by an Electromagnetic Field-Based Sensory Glove
Next Article in Special Issue
Carbon Nanomaterial-Based Electrochemical Biosensors for Alzheimer’s Disease Biomarkers: Progress, Challenges, and Future Perspectives
Previous Article in Journal
Designing the Future of Biosensing: Advances in Aptamer Discovery, Computational Modeling, and Diagnostic Applications
Previous Article in Special Issue
SERS-Driven Evolution of Lateral and Vertical Flow Assays in Medical Diagnostics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Biomarkers and Nanobiosensing Strategies in Diabetes

by
Anupriya Baranwal
1,
Vipul Bansal
1,* and
Ravi Shukla
1,2,*
1
Sir Ian Potter NanoBioSensing Facility, NanoBiotechnology Research Laboratory, School of Science, RMIT University, Melbourne, VIC 3000, Australia
2
Centre for Advanced Materials & Industrial Chemistry, RMIT University, Melbourne, VIC 3000, Australia
*
Authors to whom correspondence should be addressed.
Biosensors 2025, 15(10), 639; https://doi.org/10.3390/bios15100639
Submission received: 12 August 2025 / Revised: 17 September 2025 / Accepted: 18 September 2025 / Published: 25 September 2025
(This article belongs to the Special Issue Nano/Micro Biosensors for Biomedical Applications (2nd Edition))

Abstract

Diabetes mellitus is a chronic metabolic disorder characterised by impaired glucose regulation, leading to severe complications affecting multiple organ systems. Current diagnostic approaches primarily rely on glucose monitoring, which, while being effective, fails to capture the underlying molecular changes associated with disease progression. Emerging biomarkers such as microRNAs (miRNAs) and adipokines offer new insights into diabetes pathophysiology, providing potential diagnostic and prognostic value beyond traditional methods. Given this, precise monitoring of the altered levels of miRNAs and adipokines can forge a path towards early diabetes diagnosis and improved disease management. Biosensors have revolutionised diabetes diagnostics, with glucose biosensors dominating the market for decades. However, recent advancements in nanobiosensors have expanded their scope beyond glucose detection, enabling highly sensitive and selective monitoring of biomolecular markers like miRNAs and adipokines. These nanotechnology-driven platforms offer rapid, inexpensive, and minimally invasive detection strategies, paving the way for improved disease management. This review provides an overview of diabetes, along with its pathogenesis, complications, and demographics, and explores the clinical relevance of miRNAs and adipokines as emerging biomarkers. It further examines the evolution of biosensor technologies, highlights recent developments in nanobiosensors for biomarker detection, and critically analyses the challenges and future directions in this growing field.

1. Diabetes Mellitus: An Overview

Diabetes mellitus is a group of chronic metabolic disorders characterised by elevated blood glucose levels, commonly known as hyperglycaemia [1]. This condition arises due to defects in insulin secretion, insulin action, or both [1,2]. Insulin is a peptide hormone secreted by the pancreatic β-cells. It plays a pivotal role in regulating blood glucose by facilitating glucose uptake into cells for energy production or storage [3]. Moreover, insulin also regulates carbohydrate, lipid, and protein metabolism while promoting cell division by demonstrating mitogenic effects. According to the World Health Organisation (WHO), diabetes is a major global health issue, with an alarming increase in prevalence, particularly in low- and middle-income countries [4]. Diabetes is broadly classified into several types, with type 1 diabetes (T1D) and type 2 diabetes (T2D) being the most common forms [1,2,4].

1.1. Type 1 Diabetes (T1D)

T1D, often referred to as juvenile diabetes, is an autoimmune condition where the body’s immune system destroys insulin-producing pancreatic β-cells (Figure 1). This leads to an absolute insulin deficiency, requiring individuals to depend on exogenous insulin for survival [1,2]. The exact aetiology behind T1D remains unknown, but genetic predisposition and environmental triggers, such as viral infections, are believed to play a role [5]. Considering the multi-factor requirement to induce T1D would mean that the condition is uncommon; however, it is not rare. T1D accounts for approximately 5–10% of all diabetes cases and commonly manifests in childhood or adolescence, although it can develop at any age [1,5]. In 2021, approximately 8.4 million individuals across the globe had TID, and about 510,000 new cases were reportedly diagnosed in that year alone [6]. Of this estimate, 18% were younger than 20 years, 64% were aged between 20 and 59 years, and the remaining were 60 years or older. About 35,000 undiagnosed individuals died within one year of symptom onset. The mortality rates between countries vary greatly depending on their economic status, as 20% of individuals with T1D were in low-income and lower-middle-income countries [6].
T1D is known to develop in a clinically silent manner over a period of a few months to years. The disease progression can be divided into three stages once the commencement of autoimmunity has occurred [7,8]. Stage 1 is marked by the presence of two or more autoantibodies, stage 2 encompasses autoantibodies from stage 1 and β-cell dysfunction due to progressive loss of β-cell mass, and the clinical manifestation of T1D defines stage 3. Notably, individuals diagnosed in stage 1 exhibit a risk of 35–50% in progressing to stage 3 of T1D within 5–6 years. The risk rises to 75% for individuals being diagnosed in stage 2, with clinical manifestation of T1D in 2 years [8,9].

1.2. Type 2 Diabetes (T2D)

T2D is a complex multi-factor polygenetic disease that is primarily driven by preobesity/obesity [10]. It is characterised by insulin resistance, where the insulin-sensitive tissues fail to respond effectively to insulin (Figure 1), coupled with a progressive decline in pancreatic insulin production [11]. Consequently, elevated levels of blood glucose (hyperglycaemia) become evident in individuals with the condition. T2D primarily affects adults but is increasingly being diagnosed in younger populations due to rising obesity rates, an ageing population, and sedentary lifestyles [10]. According to the International Diabetes Federation (IDF), ~537 million adults aged 20–79 years are living with diabetes across the globe (Figure 2) [12]. If the current trends continue, this figure is expected to rise to 783 million by 2045 [13]. Of this estimate, T2D accounts for about 90–95% of cases. In 2021, high body-mass-index (BMI) was reported to contribute over 50% of the global T2D disability-adjusted life year (DALY). Accumulating reports indicate a correlation between T2D and gender difference, with males showing a higher prevalence than females [14]. Age is another important risk factor for T2D, with a rise of 24.4% in diabetes in the 75–79 years age group; however, a >1% increment was evident for the under 20 age group [15]. Other factors contributing to the prevalence of T2D include poor dietary choices, usage of tobacco, alcohol consumption, environmental factors, and physical inactivity [15]. Aside from this, genetics plays a small yet crucial role in diabetes, with an estimated heritability of 30% to 70%, depending on the age of diabetes onset and the glycaemic status of diabetic cases [16].
Globally, diabetes accounts for 12% of all health expenditures, amounting to over $966 billion USD annually [15]. This includes costs for medication, hospitalisations, and management of complications. Beyond financial costs, diabetes imposes a significant quality-of-life burden due to its chronic nature and associated complications. Efforts to mitigate the disease burden are multi-faceted, including early detection, lifestyle interventions, and advanced treatments [10]; however, healthcare disparities and the complications of diabetes management continue to pose their own challenges.

1.3. Other Types

In addition to T1D and T2D, there are other forms of diabetes, such as gestational diabetes mellitus (GDM) and monogenic diabetes [17,18]. GDM occurs during pregnancy when hormonal changes lead to insulin resistance (Figure 1), potentially affecting both maternal and foetal health [17]. While GDM usually resolves after childbirth, it increases the risk of developing T2D later in life. Monogenic diabetes, a rare form, is caused by mutations in a single gene affecting insulin production or function [18]. It includes maturity-onset diabetes of the young and neonatal diabetes. Another noteworthy type is secondary diabetes, which arises due to underlying conditions like pancreatic diseases, hormonal disorders, or medication side effects.

1.4. Secondary Complications

Prolonged hyperglycaemia can lead to several debilitating complications affecting various organ systems (Figure 3). Microvascular complications include diabetic retinopathy, which can cause vision loss, diabetic nephropathy leading to chronic kidney disease, and diabetic neuropathy, resulting in numbness, pain, or ulcers [19]. Macrovascular complications, such as cardiovascular disease, stroke, and peripheral artery disease, are common in diabetes patients and are significant contributors to morbidity and mortality [19]. Poorly managed diabetes also impairs immune function, increasing susceptibility to infections like urinary tract infections and skin disorders. Additionally, diabetic foot ulcers, often exacerbated by neuropathy and poor circulation, can lead to severe outcomes, including amputation if untreated [20]. Preventing complications requires a comprehensive approach, including regular health check-ups, strict glycaemic control, and managing comorbidities like hypertension and dyslipidaemia.

1.5. Diabetes Symptoms

The symptoms of diabetes vary depending on the type and severity of the condition. Common symptoms include polyuria (frequent urination), polydipsia (increased thirst), and polyphagia (excessive hunger) [1,4]. Unexplained weight loss, fatigue, excessive skin dryness, and blurred vision are also hallmark signs of the disease [4]. Additional signs of poorly managed diabetes include slow-healing wounds, recurrent infections, and numbness or tingling in the hands and feet due to nerve damage [4]. In GDM, symptoms are typically mild and may overlap with normal pregnancy-related changes, making routine screening critical for diagnosis.

2. Diabetes Screening and Diagnosis

Diabetes is a chronic condition characterised by persistent hyperglycaemia, which necessitates early and accurate diagnosis to facilitate timely interventions, effective disease management, and the prevention of secondary complications. The approaches of screening and diagnosis vary between T1D and T2D due to their distinct pathophysiological mechanisms and clinical presentations. For both types, biomarkers play a pivotal role in mediating early detection and risk stratification. Biomarkers, as defined by the National Institute of Health (NIH), are detectable indicators of biological processes, pathogenic conditions, or responses to interventions, and they include molecular, cellular, and physiological characteristics [21]. Their utility spans across several categories, including diagnostic, predictive, prognostic, monitoring, response, and safety biomarkers [21,22]. For the purpose of this review, however, the primary focus will be on traditional and potential diagnostic and predictive biomarkers.

2.1. T1D Screening and Diagnosis

Screening for T1D focuses on identifying individuals at risk of developing the disease before the onset of clinical symptoms. Unlike T2D, which has a gradual onset [10], T1D often presents acutely [2]. Screening efforts are typically targeted at individuals with a family history of T1D or other autoimmune disorders. The primary tools for screening include the detection of islet autoantibodies, such as glutamic acid decarboxylase antibodies (GADA), insulin autoantibodies (IAA), islet antigen-2 antibodies (IA–2A), and zinc transporter 8 antibodies (ZnT8A) [23,24,25]. These autoantibodies typically appear several months or years prior to the clinical onset and serve as predictive indicators of β-cell destruction. The presence of two or more autoantibodies in an individual is associated with a high risk of developing T1D [26]. Human leukocyte antigen (HLA) class II genotyping is another means to identify genetic susceptibility to T1D. Reportedly, specific HLA haplotypes, such as HLA DR-DQ, confer a significantly increased risk of developing T1D [27,28]. However, genetic screening is less commonly performed due to the variability in genetic predisposition and its limited predictive value. Screening is particularly relevant in research and preventive settings, as it allows for the identification of susceptible individuals who might benefit from immunotherapies or lifestyle interventions to delay or prevent disease onset. Despite these advances, routine population-wide screening for T1D has not yet been adopted due to the lack of established, widely effective preventive measures and the often sudden onset of clinical symptoms [29].
The diagnosis of T1D typically occurs when individuals exhibit symptoms of hyperglycaemia, such as polydipsia, polyuria, unintended weight loss, fatigue, and, in severe cases, diabetic ketoacidosis (DKA) [30]. DKA is a life-threatening condition characterised by the accumulation of ketone bodies in the blood, which leads to metabolic acidosis. This condition manifests as nausea, vomiting, abdominal pain, rapid breathing, and confusion [31]. Research shows that over 95% of newly diagnosed patients seek medical attention due to the presence of the aforementioned symptoms [32], while only a small percentage are diagnosed through routine glucose tests or the detection of autoantibodies as part of longitudinal screening programmes [7].
The diagnostic criteria for T1D are based on blood parameters like glucose and HbA1c levels, which reflect the glycaemic status in an individual. A fasting blood glucose (FBG) level of ≥126 mg/dL (≥7.0 mmol/L) indicates hyperglycaemia after at least 8 h without food, while a random blood glucose (RBG) level of ≥200 mg/dL (≥11.1 mmol/L) in the presence of symptoms confirms diabetes [33]. The oral glucose tolerance test (OGTT) is a strategy in which plasma glucose is measured 2 h after ingesting a glucose solution (2 h postprandial). OGTT result is diagnostic if the level reaches ≥200 mg/dL (≥11.1 mmol/L) [33], reflecting impaired glucose regulation. An HbA1c level of ≥6.5% indicates chronically elevated blood sugar levels over the previous 2–3 months but may be less specific in T1D due to its rapid onset [34]. To confirm T1D, additional tests such as C-peptide levels and autoantibody testing are often performed. Low or undetectable C-peptide levels, which measure endogenous insulin production, are indicative of β-cell failure characteristic of T1D [35,36]. Autoantibody testing helps differentiate T1D from other forms of diabetes, particularly in atypical cases or when the clinical presentation is ambiguous [25]. Early and accurate diagnosis is essential to promptly initiate insulin therapy, prevent complications like DKA, and achieve optimal glycaemic control.

2.2. T2D Screening and Diagnosis

Screening for T2D is a proactive approach aimed at identifying individuals with undiagnosed diabetes or those at high risk of developing the disease. Unlike T1D, which typically presents acutely, T2D develops slowly and often remains asymptomatic in its early stages [37]. Screening is particularly important for individuals with risk factors such as preobesity/obesity, advanced age, a family history of diabetes, or a history of gestational diabetes [10]. Preobesity/obesity is defined by body mass index (BMI), with a BMI of ≥25 kg/m2 (≥23 kg/m2 in Asia) as preobese and ≥30 kg/m2 (≥27.5 kg/m2 in Asia) as obese [38]. However, BMI alone cannot be used as an alternative for screening total fat content in individuals, as people with the same BMI may have different fat content and distribution, representing different risks of diabetes [39]. Waist-to-height ratio, defined as waist circumference (cm) divided by height (cm), has been proposed as a substitute indicator of BMI to define obesity [40]. In addition to risk factors, commonly used approaches to screen diabetes include FBG and HbA1c tests. An HbA1c level between 5.7% and 6.5%, termed prediabetes [41], reflects intermediate levels of chronic hyperglycaemia and signifies an increased risk of progression to T2D. Similarly, an FBG level between 100 and 125 mg/dL (5.6–6.9 mmol/L) indicates impaired fasting glucose, another marker of elevated diabetes risk [42]. Screening is recommended for adults aged ≥ 35 years and younger individuals with significant risk factors [37]. Regular screening enables early identification of at-risk individuals, allowing for timely lifestyle interventions or pharmacological measures to delay or prevent the onset of diabetes and its complications.
The diagnosis of T2D relies on standardised criteria similar to those used for T1D; however, distinguishing between the two types is essential due to differences in their underlying pathophysiology and management strategies. Diagnostic tests include FBG, HbA1c, RBG with symptoms of hyperglycaemia, or OGTT results showing a 2 h postprandial plasma glucose (2 h PG). The diagnostic threshold set by the American Diabetes Association (ADA) and the WHO for each criterion is captured in Table 1. These thresholds apply to both T1D and T2D, as they reflect the presence of hyperglycaemia severe enough to warrant a diagnosis of diabetes. In T2D, symptoms such as polyuria, polydipsia, and fatigue may develop gradually or be absent entirely, especially in the early stages. Recently, the IDF recognised that individuals with 1 h postprandial glucose (1 h PG) 8.6 mmol/L would be considered prediabetic, while ≥11.6 mmol/L would be diabetic [43]. Unlike T1D, C-peptide levels are typically normal or elevated in T2D due to ongoing endogenous insulin production, albeit insufficient to overcome insulin resistance [36]. Often, T2D is diagnosed incidentally during routine medical checkups or evaluations for related conditions such as hypertension, dyslipidaemia (abnormal levels of lipids in the blood), or cardiovascular disease [10].

3. Emerging Biomarkers in Diabetes

The diagnostic criteria mentioned above have established their importance in routine clinical practice due to their ease of measurement, strong correlation with long-term diabetes, and ability to guide therapeutic decisions. However, they lack the sensitivity to identify diabetes in its early or preclinical stages. This makes it difficult to judge whether changes in these parameters lead to diabetes or the onset of diabetes changes these parameters and biomarkers. In view of this, biomarkers exhibiting higher specificity and sensitivity are emerging as potential solutions to facilitate early intervention and reduce the risk of diabetes-related complications.

3.1. Emerging Biomarkers in T1D

Over the years, improved technologies for monitoring and insulin administration have significantly reduced the disease burden and improved life quality; however, T1D remains incurable. Today, when T1D is clinically established, >80% of the total β-cell mass is lost and secondary complications start becoming evident [45,46]. To prevent or delay the onset of these complications, early identification of individuals at risk of developing T1D is required, ideally prior to or independent of hyperglycaemic condition. Therefore, novel biomarkers that can identify such individuals are being searched using candidate-driven and/or screening approaches.
In the last decade, circulating microRNAs (miRNAs) have emerged as potential biomarkers for various diseases as their level gets altered with different pathological conditions [47,48,49,50]. miRNAs are short (21–24 nucleotides long) non-protein coding RNAs that play regulatory roles in post-transcriptional modifications, gene expression, and pathophysiological processes like metabolism, inflammation, and immune response [50]. Several miRNAs have been identified for their physiological involvement in tissues affected with T1D and related complications (Table 2). Their aberrant expression profile in various biological fluids, including serum, plasma, and urine, makes them accessible and minimally invasive diagnostic biomarkers. The presence of miRNAs in circulation is either to mediate inter-cellular communication or due to stressed/ruptured cells [51]. While RNAs are generally unstable and prone to degradation by nucleases, miRNAs exhibit remarkable stability in body fluids due to their association with protective carriers such as exosomes, lipoproteins, and RNA-binding proteins. These associations shield miRNAs from enzymatic degradation, making them reliable biomarkers for diagnostic applications [50]. In addition, they are resistant to repeated freeze–thaw cycles, pH fluctuations, and high temperatures, exhibit a long half-life [52] and can be accurately measured [53] from stored clinical samples.
In recent years, several other novel biomarkers have been explored, quantified, and tested for their role in predicting T1D onset and progression and risk stratification of individuals with and/or without T1D. Details on these biomarkers can be found in reviews [54,55,56,57,58,59] as their discussion falls out of the scope of this review.
Table 2. miRNA in diabetes and associated complications.
Table 2. miRNA in diabetes and associated complications.
miRNADiseases
miR-145-5pT1D [51], T2D [60,61,62], diabetic nephropathy [63], microvascular complications [64,65], macrovascular complications [66], diabetic foot ulcer [67,68], gestational diabetes [69]
miR-30e-3pT1D [70], prediabetes [71], T2D [72], microvascular complication [73], macrovascular complication [74], retinopathy [75], diabetic nephropathy [76]
miR-30c-5pT2D [77], diabetic macrovascular complication [78], diabetic nephropathy [79,80]
miR-148a-3pT1D [70,81,82], T2D [83], diabetic retinopathy [75]
miR-155-5pT1D [84,85], T2D [86], diabetic dyslipidaemia [87], diabetic foot ulcer [88]
miR-25-3pT1D [82], diabetic nephropathy [89], diabetic retinopathy [90]
miR-625-5pDiabetic cognitive impairment [91]
miR-20a-5pDiabetic foot ulcer [92], gestational diabetes [93], diabetic macrovascular complication [94,95], non-alcoholic fatty liver disease [96,97]
miR-326T1D [51,98], T2D [90]
miR-24-3pT1D [51,81,82,99], T2D [83], gestational diabetes [100], diabetic retinopathy [101,102]
miR-301b-3pT1D [51], diabetic wound healing [103]
miR-223-3pT1D [51], T2D [104], diabetic microvascular complication [105], gestational diabetes [106]
miR-21-5pT2D [83], diabetic nephropathy [89,107], diabetic foot ulcer [92]
miR-30a-5pT1D [51,82], T2D [108] and prediabetes [109], diabetic nephropathy [76,110]
miR-186-5pT1D [51], diabetic peripheral neuropathy [111], diabetic macrovascular complication [112,113], diabetic retinopathy [114]
miR-26a-5pT1D [51,82], T2D [60,83], diabetic nephropathy [89]
miR-126-3pT2D [60,83,115,116,117], diabetic retinopathy [118,119], diabetic nephropathy [76,89]
miR-199a-3pT1D [51], T2D [60], diabetic macrovascular complication [78], diabetic retinopathy [75]
miR-127-3pT1D [51,120], T2D and prediabetes [121]
let-7e-5pT1D [51,99], T2D [83] and prediabetes [121], diabetic retinopathy [75], diabetic nephropathy [107]
miR-146a-5pT1D [51,81], T2D [83,122,123] and prediabetes [124]
miR-200a-3pT1D [51,82,125], diabetic retinopathy [126], diabetic nephropathy [127,128], diabetic macrovascular complication [129]
miR-34a-5pT1D [51,130], T2D [122,124], diabetic cognitive impairment [131], diabetic cataractogenesis [132]
miR-181a-5pT1D [51,81,82], T2D [133], diabetic nephropathy [134]
miR-22-5pT1D [51], macrovascular complications [135]
miR-93-5pT1D [70], diabetic macrovascular complication [136]

3.2. Emerging Biomarkers in T2D

The increasing global burden of T2D emphasises the urgent need for precise biomarkers that can predict disease onset, monitor its progression, and provide insights into prognosis. Recent advances in molecular biology and high-throughput technologies have facilitated the discovery of novel biomarkers with significant clinical utility [137]. These biomarkers can be categorised into genetic, molecular, metabolic, inflammatory, and microbiome-related factors (Table 3). Each of these factors capture distinct aspects of T2D pathophysiology, including insulin resistance, β-cell dysfunction, and chronic inflammation. Detailed information on emerging biomarkers in T2D prediction, diagnosis, and prognosis can be found in reviews [138,139,140,141,142,143,144].
While genetic, epigenetic, and metabolomic markers have significantly advanced the understanding of T2D pathophysiology [182], adipokines offer a distinct advantage as biomarkers due to their direct contribution to the metabolic and inflammatory pathways underpinning the disease [183]. Unlike genetic and epigenetic markers, which often indicate predisposition, adipokines reflect the real-time disease status by revealing the functional state of adipose tissues. Adipokines or adipocytokines are bioactive proteins and peptides secreted by the adipose tissues. They are known to function as endocrine and paracrine factors to regulate energy homeostasis, glucose and lipid metabolism, insulin sensitivity, inflammation, and immune response [184]. The secretion patterns of adipokines are dynamically modulated by metabolic and environmental factors, which makes them highly sensitive to the systemic changes occurring in T2D [183]. Moreover, they bridge the gap between obesity and T2D by providing mechanistic insights into how adipose tissue dysfunction promotes insulin resistance and chronic inflammation [183]. Various adipokines have been implicated in T2D pathogenesis, with some exhibiting synergistic effects while others imparting antagonistic effects (Table 4). For instance, in the early stages of metabolic dysfunction, anti-inflammatory adipokines like adiponectin and vaspin may counterbalance the deleterious effects of pro-inflammatory adipokines like leptin and resistin [185]. Owing to these attributes, adipokines are emerging as not only predictive but also actionable biomarkers with potential applications in both diagnosis and personalised therapeutic strategies.
Like T1D, various circulating miRNAs have also been associated with T2D (Table 2). T2D pathogenesis is silent in nature, which develops slowly over several years and often goes unnoticed due to its asymptomatic nature. Traditional biomarkers, despite being the gold standard in T2D diagnosis, only monitor hyperglycaemia—a condition developed in the later stage of T2D pathogenesis, indicating their inadequacy in T2D prediction. In view of this, miRNAs are emerging as excellent alternatives. Reportedly, miRNA expression profiles become altered in body fluids several years prior to the clinical manifestation of T2D [50,142,203]. Therefore, accurate detection of circulating miRNAs in body fluids can help in mediating early T2D diagnosis. While this seems ideal, precise monitoring of miRNA levels is challenging due to their small size (21–24 nucleotides), sequence homology, and low abundance in biological fluids.
Over the years, a wide range of biomolecular assays have been developed and applied for the detection of miRNAs, including microarray technology [204], quantitative reverse transcription polymerase chain reaction (qRT-PCR) [205,206], rolling circle amplification (RCA) [207,208], and ligase chain reaction (LCR) [209]. While these techniques have advanced miRNA detection by offering high sensitivity and specificity, they are often constrained by inherent limitations. Microarray technology, for instance, requires labour-intensive hybridisation steps and specialised equipment [210]. Similarly, qRT-PCR, a gold standard for nucleic acid quantification, demands precise thermal cycling, expensive reagents, and skilled personnel [211], making it less accessible for widespread use. RCA and LCR, despite their innovative amplification strategies, are hindered by prolonged assay times, false positive results, and elevated costs [212]. Moreover, these approaches generally lack the flexibility and simplicity required for integration into portable, point-of-care (POC) diagnostic platforms, which emphasises the need for the development of alternative miRNA detection methods that are rapid, cost-effective, and amenable to real-time and on-site applications. In this direction, biosensors have emerged as excellent platforms for their ability to overcome the aforementioned limitations while ensuring high sensitivity and specificity. The upcoming section focuses on biosensors in diabetes by briefly touching on their historical account, their current status, and market size. Finally, it discusses the scope for new biosensor platforms in diabetes prediction and diagnosis.

4. Biosensor in Diabetes

Biosensors have revolutionised the field of diabetes diagnostics, enabling rapid, accurate, and minimally invasive monitoring of blood glucose levels. These devices have evolved from simple enzymatic systems to advanced continuous glucose monitoring technologies, addressing the growing need for effective diabetes management. A biosensor is an analytical device that converts a biological response into a measurable signal [213]. It consists of three primary components: a biomolecular recognition element (MRE), a transducer, and a signal processor (Figure 4). The MRE is often an enzyme, receptor, antibody, aptamer, or nucleic acid that specifically recognises the target analyte [214,215]. The transducer then converts the biological interaction into a measurable physical or chemical signal, such as an electrical current, thermal change, or optical response. Finally, the signal processor interprets this signal into meaningful data, which is displayed to the user. These components work together to enable rapid, accurate, and specific detection, making biosensors indispensable tools in healthcare, particularly in managing chronic conditions like diabetes.
Biosensors can typically be classified into two categories based on the type of MREs and the type of transducer [216]. MREs are the functional components that impart specificity to biosensors by selectively binding target analytes. Common types include enzymes, which catalyse reactions producing measurable signals; antibodies, which enable high-affinity binding for target antigens; and aptamers, synthetic oligonucleotides that offer chemical stability and ease of modification. Nucleic acids are widely used for detecting complementary DNA or RNA sequences. The choice of MRE directly influences sensitivity, selectivity, and operational stability, making their careful selection extremely crucial.
The classification of biosensors based on transducer results in optical (colourimetric, fluorescence, surface plasmon resonance (SPR), chemiluminescence (CL), surface-enhanced Raman scattering (SERS), electrochemiluminescence (ECL), photometric, etc.), electrochemical (potentiometric, voltammetric, photoelectrochemical, conductometric, amperometric, impedimetric, etc.), mechanical (piezoelectric, microcantilevers, etc.), and thermal (calorimetry) biosensors [216,217]. Optical biosensors detect analytes by monitoring changes in light properties, such as absorbance, fluorescence, or refractive index, resulting from specific biorecognition events [218]. Techniques like SPR and localised SPR measure refractive index shifts near a metal surface, allowing real-time, label-free detection of biomolecular interactions. Fluorescence-based sensors exploit emission intensity or lifetime changes upon target binding, while colourimetric assays rely on visible colour changes induced by nanoparticle aggregation or catalytic reactions. Additionally, SERS provides molecular fingerprinting with ultralow detection limits by amplifying Raman signals through plasmonic or semiconductor substrates [218].
Electrochemical (EC) biosensors, on the other hand, measure the electrical signals like current, voltage, or impedance, arising from the analyte-MRE interaction [219]. Among the most widely used techniques, amperometry measures current at a fixed potential, making it suitable for enzyme-based sensors such as glucose oxidase systems. Voltammetry records current as a function of applied potential to provide mechanistic insights into redox processes, while electrochemical impedance spectroscopy (EIS) monitors variations in charge transfer resistance and double-layer capacitance, enabling label-free detection of biomolecular binding events [219].
Mechanical biosensors monitor the physical changes (mass, bending, stretching, etc.) arising from binding between MRE and its cognate analyte [220]. Thermal biosensors work on the principle of measuring heat (absorption or evolution) arising from the bio/chemical reaction [217].

4.1. Historical Account of Biosensors in Diabetes

The development of biosensors for diabetes management marks one of the most significant advancements in the field of medical diagnostics (Figure 5). In 1962, the invention of the first enzymatic glucose biosensor by Clark and Lyons [221], laid the foundation for glucose monitoring technologies. This pioneering biosensor employed glucose oxidase (GOx) as the MRE to catalyse glucose oxidation and an oxygen electrode as the transducer. The transducer measured the resulting decrease in oxygen concentration levels, providing an indirect measure of glucose concentration. In 1967, building on Clark’s breakthrough work, Updike and Hicks developed the first simplified yet practical glucose biosensor by immobilising and stabilising the GOx in a polyacrylamide gel [222,223]. In 1975, the launch of the first commercial glucose biosensor for direct glucose measurement, the YSI analyser (Model 23A), was initiated by YSI Inc. This device leveraged amperometric detection of hydrogen peroxide (H2O2), where the electric current generated during the enzymatic oxidation of glucose was directly proportional to glucose concentration [224]. While H2O2 measurements could be easily achieved on miniaturised devices [225], the high operating potentials required for amperometric H2O2 measurements led to significant interference from other electroactive species present in biological samples [226]. Moreover, these biosensors were mostly limited to clinical laboratories due to their high costs. To overcome these limitations, redox mediators, such as ferrocene derivatives, thionine, methylene blue, 7,7,8,8-tetracyanoquinodimethane (TCNQ), tetrathiafulvalene (TTF), were introduced to enhance electron transfer, reduce response time, and improve sensor performance [227,228,229]. These biosensors marked the beginning of second-generation biosensors, overseeing the development of commercial screen-printed electrodes in the 1980s [227,230]. The year 1987 saw the launch of ExacTech metre—the first-ever portable EC blood glucose monitor by Medisense Inc. for glucose monitoring [231]. This breakthrough enabled diabetes patients to measure blood glucose levels at home, marking a shift from laboratory-dependent testing to patient-centric management. Of note, even today, the current operation of most commercial glucose sensors is largely similar to that of the ExacTech metre. In the 1990s, third-generation biosensors emerged, which were reagent-less and eliminated the need for redox mediators by using direct electron transfer between the enzyme and electrode [232,233]. In 1999, MiniMed Inc. launched the first commercial in vivo device that provided continuous glucose readings by relying on H2O2-mediated detection [234]. Subsequent developments focused on miniaturisation, portability, and real-time monitoring of blood glucose. Emergence of continuous glucose monitoring (CGM) systems transformed diabetes management by providing continuous data on glucose levels, alerting users to hyperglycaemia or hypoglycaemia in real-time [235,236,237]. The commercial CGM systems are predominantly EC enzyme-based devices that employ GOx chemistry and rely either on H2O2 detection or on mediated electron transfer strategies to improve performance.
In this direction, non-invasive glucose biosensor technologies have emerged with the aim of reducing patient discomfort and improving compliance [238]. These devices are commonly based on optical or transdermal approaches. The optical glucose biosensors exploit Raman spectroscopy [239], polarimetry [240], photoacoustics [241], infrared absorption spectroscopy [242], or optical coherence tomography [243] to assess the physical properties of light in the interstitial fluid or the anterior chamber of the eye. The transdermal glucose sensors sit under the skin and track changes in the interstitial glucose levels in real time. The GlucoWatch Biographer by Cygnus Inc. Reference [244] became the first transdermal glucose sensor approved by the US FDA [245]; however, it was never widely accepted in the market due to inherent shortcomings of poor accuracy, false alarm, long warm-up time, and skin irritation. Consequently, it was discontinued in 2008 [245]. Despite this setback, tremendous efforts are being made in developing non-invasive glucose monitoring systems. The Eversense system by Senseonics, for example, uses a fluorescence-based sensor implanted subcutaneously in the upper arm. The sensor is capable of transmitting glucose data for up to 180 days with minimal maintenance [246]. Clinical trials have demonstrated the system to have comparable accuracy to transcutaneous systems, with improved patient satisfaction and reduced perceived distress [247]. Another non-invasive fully implantable CGM system is designed by GlucoTrack® (Rutherford, NJ, USA) that measures glucose directly from the bloodstream. potentially offering greater accuracy and reduced lag time compared to interstitial fluid-based systems. This system potentially offers greater accuracy and reduced lag time compared to interstitial fluid-based systems [248]. These implantable technologies represent a significant advancement in CGM form factors, addressing limitations of wearability, calibration, and sensor longevity. More details on implantable non-invasive CGM systems can be found elsewhere [248,249]. Aside from this, biosensor integration with wireless and smartphone technologies has further improved their usability [250]. Modern CGM devices can transmit glucose data to smartphones, enabling patients and healthcare providers to monitor trends and adjust treatment strategies remotely.

4.2. Glucose Biosensor Market

Owing to the increasing prevalence of diabetes across the globe and the demand for more convenient and accurate glucose monitoring methods, the current glucose biosensors market is experiencing significant growth and innovation. According to various reports, the market size is estimated to be valued between USD ~10 billion to ~14 billion, with projections to reach USD 45 billion to USD 50 billion by 2030–2033 [251,252]. This expansion translates to a compound annual growth rate (CAGR) of ~8 to 13% during the forecast period and is indicative of a robust and dynamic market trajectory [251,252].
The rising global burden of diabetes is a primary catalyst for market growth. The increasing prevalence of both T1D and T2D necessitates effective and accessible glucose monitoring solutions. Moreover, growing awareness among patients and healthcare providers regarding the importance of self-monitoring blood glucose (SMBG) for optimal glycaemic control and the prevention of long-term complications is further fuelling the demand for glucose sensors [251]. Technological advancements, particularly in CGM systems, are also pushing market growth for their ability to enable real-time glucose monitoring without the need for repeated fingerstick sampling [252]. This offers patients a significant advantage in overcoming frequent discomfort from finger picking and allows ease of adherence to diabetic management plans. Miniaturisation and improved wearability of the devices are also prominent trends, as smaller devices are more discreet and easier to integrate into daily life [251]. Coupled with this is the seamless integration of smart devices, such as smartphones and smartwatches, which facilitates data sharing, remote monitoring by healthcare professionals, and personalised data analysis for improved diabetes management [251,252].
The classification of the glucose biosensor market based on devices can be broadly segmented into SMBG devices and CGM systems. SMBG represent traditional blood glucose metres requiring finger prick [253], while CGM systems form sensors that are subcutaneously inserted for continuous glucose tracking [254]. The classification based on end-users includes hospitals and clinics, home care settings, and diagnostic centres, each having specific requirements and preferences for glucose monitoring technologies. Geographically, currently, North America holds a dominant market share, attributed to a high prevalence of diabetes and the presence of major industry players within the region [251]. However, the Asia-Pacific region is anticipated to experience the most rapid growth in the coming years. This expansion is anticipated to be driven by the increasing prevalence of diabetes in populous countries like China and India, coupled with rising healthcare expenditure and improved access to advanced medical technologies.

4.3. Scope for Potential Biosensors

While glucose biosensors provide a crucial window into real-time glucose fluctuations, they offer a limited perspective on the complex interplay of metabolic processes underlying diabetes and associated conditions. Expanding the scope of biosensing to encompass other emerging biomarkers, as highlighted in Section 4, presents a significant opportunity to gain a more holistic understanding of an individual’s health status and risk of developing complications. Moreover, the integration of glucose monitoring with the assessment of other biomarkers like miRNA and adipokines offers several key advantages. Firstly, monitoring these biomarkers allows for early disease detection and more accurate risk assessment, as changes in these biomarkers precede the changes in glucose levels [50,142]. Secondly, timely interventions exhibit the potential to prevent or delay the onset of diabetes and its associated complications [51]. Moreover, monitoring these biomarkers could facilitate the development of personalised treatment strategies based on the patient’s unique metabolic profile and risk factors. By providing a deeper understanding of the underlying disease mechanisms, these biomarkers can also contribute to the development of novel therapeutic targets [50,51]. Lastly, monitoring these biomarkers exhibits the potential to improve long-term complications prediction, allowing for more proactive management and improved patient outcomes [50]. In view of this, expanding the biosensing technology beyond glucose to include emerging biomarkers like microRNA and adipokines offers a more comprehensive and nuanced approach to diabetes management, moving towards a future of personalised and preventive medicine.

5. Nanobiosensors in Diabetes

The amalgamation of sensor technology with nanotechnology has played an important role in developing robust detection systems. These biosensors utilise nanomaterials, including but not limited to metal-based, metal oxide-based, quantum dots (QDs), carbon-based, and metal–organic frameworks (MOFs), to confer high sensitivity, specificity, and detection versatility [255]. Nanomaterials are small materials or chemical substances that possess sizes in the range of 1–100 nm for at least one dimension. They can be classified into different categories based on their morphology, size, composition, and dimensions [256]. The nano-dimensional size of nanomaterials endows them with unique physicochemical properties, such as optical, electrical, thermal, and mechanical characteristics, that are typically absent in bulk materials. The ability of nanomaterials to possess a high surface area-to-volume ratio combined with tunable shape and size and ease of surface functionalisation places nanobiosensors in a different league from conventional biosensors, as these properties enable nanobiosensors with high sensitivity and flexible detection capabilities [255].

5.1. Nanobiosensors in miRNA Detection

Over the years, myriad reports investigating the potential of nanobiosensors in miRNA detection have been published (Figure 6). These biosensors commonly employ single-stranded DNA (ssDNA) complementary to the target miRNA as the MRE and utilise nanomaterials with varying shapes, sizes, and compositions to obtain excellent sensitivity and signal amplification. While different detection modalities, including EC, fluorescence, SERS, ECL, photothermal, CL, SPR, and colourimetry, have been developed for miRNA detection (Table 5), most of them transduce the ssDNA-target miRNA hybridisation event. Considering the immense potential of (nano)biosensors in miRNA detection, various reviews have been published in recent years. While some of these reviews present a general overview of different biosensors in miRNA detection [257,258,259,260,261], others specifically focus on either a specific (nano)material type, e.g., QDs [262], magnetic nanoparticles [263], MOFs [264], the specific type of miRNA [265], or the mode of detection, e.g., plasmon-enhanced [266], electrochemiluminescence [267], electrochemical [268,269], and visual [270] biosensors. For the purpose of the review, a brief discussion on different types of nanobiosensors in miRNA detection is presented below.

5.1.1. EC Nanobiosensors in miRNA Detection

With the success of commercial portable glucose metres, EC biosensors have established their potential as simple, affordable, rapid, and user-friendly platforms. Expectedly, EC biosensors have seen tremendous growth for other targets as well, with miRNAs gaining tremendous attention in the recent past [265,291]. Much like other EC biosensors that measure changes in EC signals arising from bio/chemical events at the electrode surface, the EC miRNA biosensor monitors the hybridisation event between the immobilised ssDNA and cognate target miRNA [268]. Upon hybridisation, change in the EC signal is monitored using techniques like cyclic voltammetry (CV), differential pulse voltammetry (DPV), or EIS [268]. These EC readouts differ in how they probe the electrode interface and thus in their suitability for miRNA assays. CV sweeps the electrode potential and records oxidation/reduction currents, providing mechanistic information about redox-active reporters and surface reaction kinetics, but with modest sensitivity [219]. DPV and square-wave voltammetry (SWV) employ superimposed pulses that suppress capacitive background current, yielding high sensitivity for trace analytes. This makes them methods of choice when a redox reporter or intercalator (e.g., methylene blue, ferrocene) is used for miRNA readout. EIS applies a small alternating current across a range of frequencies to measure interfacial properties such as charge-transfer resistance and capacitance. This technique is particularly valuable for label-free hybridisation assays, as target binding events induce measurable changes in the impedance spectrum, albeit at the cost of longer measurement and more complex data fitting. In practice, CV is primarily a characterisation tool, DPV/SWV are favoured for sensitive, label-based detection, and EIS is preferred for label-free, surface-sensitive miRNA assays. Merits of nanomaterials combined with advancements in nanotechnology have led to the development of single- [292,293] and dual-mode [271,272,273] EC biosensors with sensitivities comparable to gold-standard real-time RT-PCR while maintaining high specificity and rapid turn-around time. In addition, recent efforts have transitioned away from validating only proof-of-concept demonstrations and are focusing on enhancing the sensitivity and target selectivity required to meet clinical standards. The schematics of EC nanobiosensor fabrication using single- and dual-mode miRNA detection are displayed in Figure 7.
Notwithstanding the short duration of miRNAs being considered as potential diagnostic and prognostic biomarkers, EC biosensors have undergone remarkable progress; however, numerous challenges persist. While reports on multiplex miRNA detection have started emerging, most studies commonly focus on single miRNA detection (Table 5), often miR-21, by either synthesising new nanomaterial for electrode modification or signal amplification or combining different amplification techniques to improve biosensor sensitivity [291]. The incorporation of amplification techniques results in higher sensitivity but also contributes to increasing the overall cost, detection time, and complexity of the platform [257]. Specific details on the different target miRNAs tested using electrochemical biosensor and their association with specific health conditions are presented in Table 5. Moreover, nanomaterials used in electrode modification, detection modality (single, dual, or multi-mode), biosensor analytical performance, and validation in simulated/real-clinical samples are also compiled within Table 5.

5.1.2. Optical Nanobiosensors in miRNA Detection

A wide range of optical nanobiosensors, like fluorescence, CL, ECL, SPR, colourimetric, and SERS, have been applied to miRNA detection [218]. These platforms rely on light-based signal transduction but differ in their underlying principles. Fluorescence biosensors detect changes in light emission from fluorophores or nanomaterials. They commonly operate through mechanisms such as fluorescence resonance energy transfer (FRET), where energy is transferred between a donor and an acceptor in close proximity, or through photoinduced electron transfer (PET) and aggregation-induced emission (AIE), depending on the sensor design. CL produces light through chemical reactions, while in ECL, the luminescent species are activated electrochemically at an electrode surface, enabling precise control of signal generation. SPR measures changes in the refractive index near a metal surface when biomolecules bind at the surface, whereas colourimetric biosensors use visible shifts in solution colour, typically from nanoparticle aggregation or dispersion. SERS, in contrast, detects molecular binding events through the enhancement of Raman scattering by plasmonic nanostructures, offering highly specific molecular “fingerprints” with strong sensitivity.
Fluorescence nanobiosensors typically rely on fluorescence properties of nanomaterials such as QDs, carbon dots (CDs), graphene oxide (GO), gold nanoparticles (AuNPs), and upconverting nanoparticles (UCNPs), which either act as fluorescent signal emitters or quenchers [265,294]. The mechanism often involves fluorescence resonance energy transfer (FRET), photoinduced electron transfer (PET), or aggregation-induced emission (AIE), depending on the choice of nanomaterial and detection approach [294]. For miRNA detection, these biosensors often employ a hybridisation-based approach, where fluorescently labelled probe DNAs complementary to the target miRNA are immobilised onto or interact with the nanomaterial [257,265]. Upon hybridisation, changes in fluorescence signal occur commonly due to mechanisms like FRET, where energy transfer between the donor and acceptor fluorophores or nanomaterials indicates the presence of the target miRNA. AuNPs [277,279] and GO [295] (Figure 8a) have been widely explored for their quenching potential due to their high surface area and ability to absorb fluorescence energy, creating a “turn-off” state. In the presence of the target miRNA, the fluorescence signal recovers due to hybridisation, creating a “turn-on” state. QDs [276] (Figure 8b) and CDs [296] are gaining popularity in fluorescence biosensor platforms due to their tunable emission spectra and excellent photostability over organic dyes [294]. By employing nanomaterials in fluorescence biosensors, sensitivities ranging from a few nanomolar to femtomolar have been achieved for miRNA detection (Table 5). While some studies employed amplification strategies [279,282,284,285,295] to higher sensitivity, others could achieve similar sensitivity without it [275,276,277,278,280,281,283]. Moreover, excellent recoveries could be obtained in spiked biological samples, indicating their potential for practical application. Specific details on the target miRNA and their implication in particular health conditions, nanomaterials used in biosensor fabrication, mode of detection, biosensor analytical performance, and their validation in simulated/real-clinical samples are discussed in Table 5.
CL and ECL biosensors have emerged as powerful tools for miRNA detection, offering distinct advantages over traditional fluorescence-based methods [257]. These techniques rely on generating light through chemical or EC reactions, eliminating the need for an external light source and reducing background interference. In CL biosensors, the reaction between a chemical species (e.g., luminol) and an appropriate catalyst produces light emission, which is coupled with miRNA recognition events through amplification strategies [285,297] and/or enzyme-linked reactions [298] (Figure 9). ECL, on the other hand, involves the generation of light through electrochemical stimulation of a luminescent species (e.g., luminol, QDs) at an electrode surface [274,299]. This technique offers higher sensitivity (aM) and control over the reaction compared to CL, as the light emission can be precisely triggered by applying a potential [274]. ECL biosensors can be designed with various configurations, including direct detection of miRNA hybridisation on the electrode surface or indirect detection using labelled probes or nanoparticles [267]. Both CL and ECL-based sensors offer several advantages over fluorescence biosensors. CL and ECL exhibit lower background noise and autofluorescence, i.e., superior signal-to-noise ratio due to the absence of an external light source, leading to improved sensitivity [257]. Furthermore, they circumvent issues associated with photobleaching, thereby resulting in more stable and reliable signals [267]. Photobleaching is a common problem in fluorescence assays that tends to significantly reduce signal intensity over time. However, it should be noted that photobleaching is principally a concern for applications involving repeated or continuous optical excitation and is less problematic for single-use point-of-care assays. Conversely, while CL/ECL overcome light-induced fluorophore degradation, they can face other long-term stability issues like luminophore degradation, electrode fouling, and loss of EC efficiency for long-term or continuous applications. Details on the target miRNAs tested by CL and ECL approach in combination with other approaches, their relation to specific health conditions, nanomaterials used in biosensor fabrication, analytical metrics, and their validation in simulated/real-clinical samples are discussed in Table 5.
Colourimetric biosensors are another type of optical biosensor that have found their application in miRNA sensing due to their simplicity and cost-effectiveness. These biosensors utilise visible colour changes to indicate the presence or absence of target miRNA. The most common strategies for colourimetric miRNA detection involve AuNPs [300], enzyme-based assays [301,302], and nanozyme-mediated [286,288] signal transduction. AuNP-based approaches exploit localised surface plasmon resonance (LSPR) of nanoparticles to enable dramatic shifts in colour in response to aggregation or dispersion [300]. Typically, ssDNA probes complementary to the target miRNA are immobilised onto AuNPs [303]. In the absence of the target miRNA, the AuNPs remain dispersed, retaining their red colour. However, in the presence of target miRNA, hybridisation-induced conformational change or salt-induced aggregation occurs [303,304]. This causes a distinct shift in colour from red to purple or blue, providing a visual readout (Figure 10a). Enzyme-mediated systems employ natural peroxidases like horseradish peroxidase (HRP) to catalyse the oxidation of peroxidase substrates, 3,3’,5,5’-tetramethylbenzidine (TMB) and hydrogen peroxide (H2O2), to produce visual colour when target miRNA is present (Figure 10b) [301,302]. The biosensor specificity is often achieved by employing ssDNA probes, which hybridise with the target miRNA to induce the desired visual change. Nanozyme-based assays employ nanomaterials exhibiting intrinsic enzyme-mimic catalytic activity to generate a visual signal response [305] (Figure 10c). The incorporation of nanozymes in biosensor platforms has gained momentum for their obvious merits over natural enzymes. Enzymes, being proteins in nature, are prone to denaturation and deactivation under ambient conditions, suffer from expensive and complex production and purification procedures, and are restricted to a narrow temperature and pH range for optimal performance [305,306]. In contrast, nanozymes offer several advantages, including higher stability, tolerance to wider temperature and pH ranges, lower production cost, and catalytic activity similar to, if not better than, natural enzymes [306]. Despite these merits, however, several challenges limit their translation into clinical diagnostics. Unlike natural enzymes, nanozymes often lack substrate specificity, and their catalytic activities can be influenced by non-biological factors, reducing their reliability in complex clinical samples. Moreover, nanozyme activity is highly dependent on synthesis conditions, raising concerns about batch-to-batch reproducibility and scalability for regulatory approval. Thus, while nanozymes show strong promise in proof-of-concept studies, their current performance as catalytic materials has not yet been translated outside controlled laboratory environments, and further work is required to enhance their specificity and clinical reliability.
Colourimetric biosensors, in general, have been reported to achieve nanomolar [307] to picomolar [286] sensitivities for miRNA detection in the absence of amplification techniques. While these assays are promising for their simple and easy-to-use setup, they lack the sensitivity required to meet clinical standards. Despite undergoing overexpression under disease conditions, miRNA level is known to remain within the femtomolar to attomolar range. In view of this, amplification techniques like strand displacement amplification (SDA) [308], HCR [288], HCR with DNAzyme [287], and rolling circle amplification (RCA) with DNAzyme [309] have been utilised to obtain femtomolar to attomolar [288] sensitivity. Recently, efforts have also been directed towards developing sensors that can achieve desirable sensitivity while being enzyme-free as well as amplification-free [310,311]. Specific details on the target miRNA tested using colourimetric nanobiosensors, along with nanomaterials used in sensor fabrication, analytical performance, and their validation in simulated/real-clinical samples, are discussed in Table 5.

5.2. Nanobiosensors in Adipokine Detection

T2D is a chronic metabolic disorder that requires early diagnosis and monitoring to prevent the development of severe complications like cardiovascular diseases, neuropathy, retinopathy, and nephropathy. While conventional diagnostic biomarkers highlighted in Section 3.2, like FPG, RBG, 2 h PG, and HbA1c, are widely used, they often fail to provide real-time insights into metabolic dysfunction at an early stage. To this end, adipokines have emerged as promising biomarkers for T2D due to their direct contribution to insulin signalling, inflammation, and energy homeostasis. Over the years, conventional techniques, such as mass spectrometry and ELISA, have been used to measure adipokine levels. Despite offering high sensitivity and specificity, these techniques are limited by their requirements for exhaustive protocols, expensive reagents, trained personnel, sophisticated instruments, and designated laboratory space. Given this, nanobiosensors have emerged as excellent alternatives due to their rapid, highly sensitive, cost-effective, and user-friendly approach to adipokine detection. The high specificity is ensured by using antibodies or aptamers as MREs.
In recent years, nanomaterials, including metal nanostructures (e.g., AuNPs [312,313,314], gold nanotriangles (AuNTs) [315], gold nanorods (AuNRs) [316], silver nanowires (AgNWs) [317]), metal oxide nanostructures (e.g., zinc oxide nanorods (ZnO NRs) [318], zinc oxide nanoparticles (ZnO NPs) [319], iron oxide nanoparticles (Fe3O4/Fe2O3 NPs) [320], titanium dioxide nanoparticles (TiO2NPs) [321]), bimetal nanoparticles (e.g., iron-nickel nanoparticles (FeNi NPs) [322], Fe3O4 core and Au shell (Fe3O4@Au) [323], Au core and Ag-Au alloy shell (Au@Ag-Au) [324]), carbon-based nanomaterials (e.g., reduced GO (rGO) [325], graphene [326]), and others (e.g., MOFs [327], UCNPs [328], etc.) have been employed in developing biosensors for adipokine detection. So far, detection modalities, such as EC, colourimetric, fluorescence, SERS, SPR, CL, photothermal, etc., have been explored for adipokine detection; however, EC remains the most exploited mode. The following section focuses on recent developments in nanobiosensors for adipokine detection using the aforementioned modes.

5.2.1. EC Nanobiosensors in Adipokine Detection

Owing to their inherent sensitivity, EC nanobiosensors have emerged as powerful analytical platforms for adipokine detection. The operational principles of EC biosensors are diverse and are often tailored to the specific requirements of the target adipokine and the intended application. The amperometric detection, for instance, is suitable when the analyte or a reporter molecule undergoes a redox process, facilitating rapid electron exchange and yielding real-time data [329]. Voltammetric techniques are ideal for detecting subtle changes in the EC profile induced by adipokine binding events, particularly when the sensor surface is modified with nanomaterials to promote electron transfer [319,325,326,330]. Meanwhile, in the EIS method, the binding of an adipokine typically leads to alterations in charge transfer resistance and capacitance, enabling detection at ultra-low concentrations [321,322,331]. To confer adipokine specificity in these EC biosensors, antibodies have long served as the primary MRE due to their inherent high affinity and selectivity for target adipokine. These sensors utilise immobilised antibodies on the electrode surface, where measurable EC changes are observed in response to the specific adipokine binding. For instance, antibody-functionalised electrodes have been successfully employed for the sensitive detection of leptin [322,325,326,330] and TNF-α and IL-6 [329] simultaneously. Combining nanomaterials with antibodies ensured fg/mL LOD towards leptin (Figure 11a), while pg/mL LOD was obtained for TNF-α and IL-6. Despite the obvious merits of antibodies, their imitations of poor stability and high production and purification costs have led to the development of alternative MREs. One such example is aptamers, single-stranded DNA or RNA oligonucleotides that fold into unique three-dimensional structures to bind with their targets. Being commonly DNA in nature, these aptamers exhibit significant advantages over antibodies, such as high stability, inexpensive synthesis, and ease of tunability, while exhibiting high target specificity. For instance, aptamer-functionalised electrodes have been successfully employed for leptin [319,321] (Figure 11b) and IL-6 [331] detection, yielding fg/mL and pg/mL detection limits for leptin and IL-6, respectively. Table 6 compiles examples of various adipokines detected using EC nanobiosensors and highlights their specific details, including health condition, nanomaterial used in electrode modification, sensor analytical performance, and MRE. It also provides information on the practical applicability of the biosensor platform.
While EC nanobiosensors exhibit promising attributes for adipokine detection, several critical limitations hinder their widespread clinical and research adoption. One significant challenge is the reproducibility of sensor fabrication arising from batch-to-batch variability in nanomaterial synthesis. This can lead to inconsistent sensor performance and compromise its quantitative accuracy. The complexity of biological matrices such as serum or plasma presents issues of non-specific adsorption and interference from other electroactive species, which may result in increased background noise and reduced specificity. The long-term stability and durability of these sensors also remain problematic, as electrode fouling and the degradation of MREs can limit sensor lifespan and operational reliability.

5.2.2. Optical Nanobiosensors in Adipokine Detection

In recent years, optical detection modalities, including colourimetric, fluorescence, SERS, CL, and SPR, have been investigated for adipokine detection. Colourimetric nanobiosensors for adipokine detection operate by inducing a visible colour change upon their binding, often through AuNP aggregation [312,314], enzyme-linked [335], or nanozyme-based [327,333] reactions. In the aggregation-based approach, AuNPs are functionalised by adipokine-specific aptamers using covalent [314] or non-covalent [312] interactions. In the presence of target adipokine (RBP-4), the non-covalently bound aptamers desorb from the AuNP surface, leaving the nanoparticle exposed to salt-induced aggregation, hence the visible colour change [312] (Figure 12a). In the case of covalent functionalisation, aptamer pairs are used to form a sandwich format and the presence of adipokine (IL-6) is confirmed by the change in colour due to AuNP aggregation [314]. The enzyme-based method also employed a sandwich format; however, it used antibody-pair, i.e., capture-detection antibodies. The capture antibodies were functionalised onto magnetic beads, while detection antibodies were labelled with HRP. In the presence of target IL-6, a sandwich formation occurs, enabling HRP to oxidise the TMB substrate using H2O2, which is further oxidised to produce visible colour using H2SO4 [335]. The nanozyme-based system employs a similar approach for IL-6 detection with minor changes [333]. In this case, the HRP enzyme was replaced with peroxidase-mimic Pd@Pt nanodendrites (Pd@Pt NDs) (Figure 12b), and the rest of the assay was kept the same. Table 6 compiles examples of various adipokines detected using colourimetric nanobiosensors and highlights their specific details, including health conditions, nanomaterials used as labels or signal amplifiers, sensor analytical performance, and MRE. It also provides information on the practical applicability of the biosensor platform.
While colourimetric nanobiosensors are simple, user-friendly, and instrument-free, their wider application is often restricted due to their limited sensitivity. Given this, fluorescence nanobiosensors are being developed for adipokine detection. These biosensors rely on the emission of light from UCNPs [328], carbon QDs [332] or fluorescent tags [318] to enable vaspin (Figure 13a), IL-6, and TNF-α detection, respectively. Despite using different MREs, aptamers for vaspin and antibodies for TNF-α, both sensors relied on the sandwich format to mediate adipokine target detection, enabling sensitivities in the range of pg/mL to fg/mL. FRET-based detection is another approach to mediate adipokine detection, wherein the sensor operates on the FRET on/off mechanism by utilising aptamer as MRE and carbon QDs and AuNPs as donor-quencher pair [332] (Figure 13b). In the absence of target IL-6, AuNPs functionalised with aptamers quench the QD fluorescence by FRET, rendering them in ‘FRET on’ the state. However, no such quenching is evident in the presence of the target due to the increased distance between the donor and quencher pair, resulting in a ‘FRET off” state. More details of these examples, including the implication of adipokine in specific health conditions, nanomaterials employed in sensor development, analytical performance metrics, and type of MRE, are summarised in Table 6. Moreover, information on the practical applicability of the biosensor platform is also highlighted depending on whether sensor performance was validated in real biological samples. Albeit fluorescence biosensors hold tremendous potential, only a handful of reports have emerged in adipokine detection, which indicates the need for extensive efforts in this direction. Moreover, most of these adipokines are associated with health conditions beyond diabetes, which, while being useful, cannot be directly expected to accurately analyse adipokines in diabetic samples as their levels tend to vary depending on disease conditions.
Other less commonly explored optical detection modes in adipokine sensing include SPR and SERS. SPR-based biosensors rely on the principle of plasmonic resonance, where changes in refractive index at the sensor surface alter the resonance angle of incident light due to biomolecular interactions. Recently, AuNRs [316] and magnetic nanoparticles (MNPs) [320] have been exploited for their application in SPR biosensors for adipokine detection. The AuNR-based sensor utilised adipokine-specific antibodies to enable simultaneous detection of TNF-α, IL-6, and TGF-β with pg/mL sensitivity [316]. In contrast, the MNP-based sensor detected RBP-4 along with other cytokines, neutrophil gelatinase-associated lipocalin (NGAL) and interleukin-18 (IL-18) with ng/mL sensitivity, using antibody-pair in a sandwich format [320]. Apart from the simultaneous detection of three clinically relevant biomarkers, both sensors displayed potential for practical applicability by detecting targets in real biological matrices. More details of these examples, including the implication of adipokine in specific health conditions and biosensor analytical performance, are summarised in Table 6.
SERS nanobiosensors offer ultrasensitive adipokine detection by exploiting the plasmonic enhancement of Raman signals by metal nanostructures such as AuNTs [315] and Au@Ag-Au core–shell [324] nanoparticles. The underlying detection principle of these biosensors was based on the amplification of vibrational spectra due to the adsorption of adipokine onto these plasmonic substrates, thereby enabling label-free detection of adiponectin [315] and IL-6 [324] with fg/mL sensitivity. In both biosensors, high specificity was conferred by the functionalisation of the nanostructure surface with antibodies, while Raman reporter molecules facilitated the acquisition of distinct spectral fingerprints upon adipokine binding. More details of these examples, including the implication of adipokine in specific health conditions and biosensor analytical performance, are summarised in Table 6.
Despite the tremendous potential of SPR and SERS-based nanobiosensors in adipokine detection, there remains a significant lack of efforts to detect these biomarkers. This lacuna can potentially be attributed to the challenges associated with the respective platforms. For instance, both SPR- and SERS-based biosensors may suffer from poor sensitivity in analysing trace analytes in complex biological matrices and may undergo non-specific binding, compromising sensor performance [336]. Another potential limitation of these biosensors is that MREs may lose their native configuration during immobilisation on the sensor chip surface, or their orientation may sterically hinder the analyte binding [336]. Similarly, SERS nanobiosensors can suffer from reproducibility issues due to batch-to-batch variability in nanomaterial fabrication [337]. In complex biological matrices, nanomaterials may undergo undesirable morphological changes due to aggregation or non-specific biomolecule adsorption, causing loss of signature SERS signals [337]. Additionally, both platforms may experience difficulties in maintaining sensor stability and specificity in heterogeneous samples and face challenges related to surface fouling and surface functionalisation required to ensure selective adipokine binding. These limitations highlight the need for continued optimisation in nanofabrication techniques and surface chemistry to fully realise the potential of SPR and SERS in clinical adipokine diagnostics.

6. Challenges and Future Perspectives

Although nanobiosensor technologies have significantly advanced the detection of circulating miRNAs and adipokines, several critical challenges must be addressed to translate these innovations into clinical practice for diabetes diagnosis and monitoring.
Currently, the majority of nanobiosensor platforms target miRNAs associated with oncological diseases, with limited efforts directed toward diabetes-related miRNAs. Furthermore, most studies focus on the detection of individual miRNAs, such as miR-21 or miR-155, while only a limited number investigate the multiplex detection of miRNAs within a unified sensor platform. It is crucial to highlight that individual miRNAs, such as miR-21, may be implicated in various disease conditions, thereby rendering the detection of a single miRNA insufficient for definitive diagnosis of any particular health condition. The development of multiplexed biosensing platforms capable of detecting panels of diabetes-specific miRNAs and adipokines simultaneously is essential for improving diagnostic accuracy and capturing the complex biomolecular signatures associated with disease progression.
Under pathological conditions, miRNA levels undergo either over- or under-expression, yet their circulating concentrations generally fall within the femtomolar to attomolar range. Consequently, amplification strategies such as RCA, SDA, and HCR are employed to enhance sensitivity. While these techniques improve detection capabilities, they inadvertently increase assay complexity, response time, and overall costs, which counteracts the primary objective of developing simple, rapid, and affordable biosensor platforms.
In the domain of adipokine detection, current efforts predominantly rely on antibody-based biosensors integrated into EC platforms. While antibodies offer excellent target specificity, their inherent instability under ambient conditions and high production costs pose substantial limitations. Antibody-derived fragments, including single-chain variable fragments (ScFv) and antigen-binding fragments (Fab), represent promising alternatives. These fragments retain high target specificity while offering advantages such as smaller size, which can improve target accessibility and reduce steric hindrance, and potentially enhanced stability under various conditions. Despite these benefits, challenges remain in their consistent large-scale production, reliable functionalization, and long-term stability, which must be addressed to enable their practical implementation in next-generation biosensing platforms. Aptamer-based biosensors provide a promising alternative with advantages in stability, cost, and ease of synthesis; however, only a limited number of adipokine-specific aptamers have been developed, and their integration into nanobiosensing platforms for diabetes biomarkers remains in its infancy.
Beyond analytical performance, manufacturability represents a major bottleneck in the clinical translation of these nanobiosensors. The ability to design sensors with consistent quality across batches is essential for achieving reliable calibration, securing regulatory approval, and ensuring reproducible clinical performance. Many promising sensors remain confined to proof-of-concept demonstrations because they cannot be manufactured reliably at scale, which ultimately limits their clinical utility. Incorporating design-for-manufacturing principles early in development, such as standardised fabrication processes, robust material selection, and scalable assembly, can significantly enhance the likelihood of successful translation.
Future research should focus on expanding the repertoire of diabetes-specific miRNAs and adipokines targeted by nanobiosensors, alongside the development of robust multiplex detection platforms. Innovative signal amplification strategies that maintain simplicity and cost-effectiveness must be prioritised to enhance sensitivity without compromising practicality. Furthermore, the exploration of enzyme-free biosensors, such as those utilising nanozymes, offers a promising avenue, though this field is still in its early stages. Significant work is also needed to improve the clinical validation of these technologies through large-scale studies using real patient samples, ensuring reliability and reproducibility in diverse clinical settings.
Ultimately, overcoming these challenges will require a multidisciplinary approach that bridges advances in materials science, molecular biology, and clinical diagnostics. The successful development of next-generation nanobiosensors capable of detecting miRNAs and adipokines with high sensitivity, specificity, ease-of-use, and manufacturability has the potential to transform the early diagnosis and personalised management of diabetes and its complications.

Author Contributions

Conceptualization, R.S. and A.B.; writing—original draft preparation, A.B.; writing—review and editing, R.S., V.B. and A.B.; supervision, R.S. and V.B.; funding acquisition, V.B. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Australian Research Council for funding support through the Discovery scheme (DP230101650); Leona M. and Harry B. Helmsley Charitable Trust (Grant 2018 PG-T1D009); the JDRF Australia Type 1 Diabetes Clinical Research Network (Grant 3-SRA-2019-694-M-B).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were generated in this study. All data used in this review are publicly available via the references provided in the text.

Acknowledgments

A.B. acknowledges the Australian government and RMIT University for the RTP Stipend Scholarship towards a PhD degree. The authors acknowledge the Ian Potter Foundation for establishing the Sir Ian Potter NanoBioSensing Facility at RMIT University, which continues to enable us to develop next-generation detection technologies.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

1,5-AHG1,5-anhydroglucitol
2 h PG2 h postprandial plasma glucose
ADAAmerican Diabetes Association
Ag/Pt NCsAg/Pt nanoclusters
AgNWsSilver nanowires
AIEAggregation-induced emission
ALPAlkaline phosphatase
AuNPsGold nanoparticles
AuNPsGold nanorods
AuNP-VS4 NS NRAuNP-vanadium sulphide nanosheet self-assembled nanorod
AuNTsGold nanotriangle
BCCAsBranched-chain amino acids
BMIBody-mass-index
CAChronoamperometry
CAGRCompound annual growth rate
CDsCarbon quantum dots
CGMContinuous glucose monitoring
CHACatalytic hairpin assembly
CPCarbon paper
CRETChemiluminescence resonance energy transfer
CRPC-reactive protein
CVCyclic voltammetry
DALYDisability-adjusted life year
DKADiabetic ketoacidosis
DPVDifferential pulse voltammetry
ECElectrochemical
ECLElectrochemiluminescence
EISElectrochemical impedance spectroscopy
ELISAEnzyme-linked immunosorbent assay
Fe3O4 NPsIron oxide nanoparticles
Fe-Ni NPsIron-nickel nanoparticles
FGF-21Fibroblast growth factor-21
FPGFasting plasma glucose
FRETFluorescence resonance energy transfer
GADAGlutamic acid decarboxylase antibodies
GDMGestational diabetes mellitus
GNPsGold nanoparticles
GOGraphene oxide
GOxGlucose oxidase
H2O2Hydrogen peroxide
HbA1cGlycated haemoglobin
HCRHybridisation chain reaction
HDLHigh-density lipoprotein
HLAHuman leukocyte antigen
HRPHorseradish peroxidase
IA-2AIslet antigen-2 antibodies
IAAInsulin autoantibodies
IDFInternational Diabetes Federation
IL-6Interleukin-6
iSDAIsothermal strand displacement amplification
LCRLigase chain reaction
LDLLow-density lipoprotein
LFDLateral flow device
LODLimit of detection
LSPRLocalised surface plasmon resonance
MBMethylene blue
MCH6-mercapto-1-hexanol
MCP-1Monocyte chemoattractant protein-1
miRNAmicroRNA
MNPsMagnetic nanoparticles
MOFsMetal–organic frameworks
MREBiomolecular recognition element
MWMethylene white
NCQNitrogen-doped carbon quantum dots
NGALNeutrophil gelatinase-associated lipocalin
NIHNational Institute of Health
NRNot reported
OCVOpen circuit voltage
OGTTOral glucose tolerance test
Pd@Pt NDs Pd@Pt core@shell nanodendrites
PETPhotoinduced electron transfer
POCPoint-of-care
PPYPolypyrrole
PtNP-Ce-MOFPtNP-modified Cerium MOF
QDsQuantum dots
qRT-PCRQuantitative reverse transcription polymerase chain reaction
RBP-4Retinol binding protein-4
RCARolling circle amplification
rGOReduced graphene oxide
RPGRandom plasma glucose
SDAStrand displacement amplification
SERSSurface-enhanced Raman scattering
SHBGSex hormone-binding globulin
Si QDsSilicon quantum dots
SMBGSelf-monitoring blood glucose
SPCEScreen-printed carbon electrode
SPEScreen-printed electrode
SPRSurface plasmon resonance
SWVSquare wave voltammetry
ssDNASingle-stranded DNA
T1DType 1 diabetes
T2DType 2 diabetes
TCNQ7,7,8,8-tetracyanoquinodimethane
TFOTriplex-forming oligonucleotide
TiO2NPsTitanium dioxide nanoparticles
TMB3,3’,5,5’-tetramethylbenzidine
tMBTriplex molecular beacon
TNF-αTumour necrosis factor-α
TTFTetrathiafulvalene
UCNPsUpconverting nanoparticles
WHOWorld Health Organization
ZnO NPsZinc oxide nanoparticles
ZnO NRsZinc oxide nanorods
ZnT8AZinc transporter 8 antibodies

References

  1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2013, 36 (Suppl. S1), S67–S74. [Google Scholar] [CrossRef]
  2. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2010, 33 (Suppl. S1), S62–S69. [Google Scholar] [CrossRef] [PubMed]
  3. Wilcox, G. Insulin and insulin resistance. Clin. Biochem. Rev. 2005, 26, 19. [Google Scholar]
  4. World Health Organization. Diabetes: Overview and Key Facts; World Health Organization: Geneva, Switzerland, 14 November 2024; Available online: https://www.who.int/news-room/fact-sheets/detail/diabetes (accessed on 3 December 2024).
  5. Deshpande, A.D.; Harris-Hayes, M.; Schootman, M. Epidemiology of diabetes and diabetes-related complications. Phys. Ther. 2008, 88, 1254–1264. [Google Scholar] [CrossRef]
  6. A Gregory, G.; Robinson, T.I.G.; E Linklater, S.; Wang, F.; Colagiuri, S.; de Beaufort, C.; Donaghue, K.C.; Magliano, D.J.; Maniam, J.; Orchard, T.J.; et al. Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: A modelling study. Lancet Diabetes Endocrinol. 2022, 10, 741–760. [Google Scholar] [CrossRef] [PubMed]
  7. Katsarou, A.; Gudbjörnsdottir, S.; Rawshani, A.; Dabelea, D.; Bonifacio, E.; Anderson, B.J.; Jacobsen, L.M.; Schatz, D.A.; Lernmark, Å. Type 1 diabetes mellitus. Nat. Rev. Dis. Primers 2017, 3, 17016. [Google Scholar] [CrossRef]
  8. Herold, K.C.; Delong, T.; Perdigoto, A.L.; Biru, N.; Brusko, T.M.; Walker, L.S. The immunology of type 1 diabetes. Nat. Rev. Immunol. 2024, 24, 435–451. [Google Scholar] [CrossRef]
  9. Evans-Molina, C.; Sims, E.K.; DiMeglio, L.A.; Ismail, H.M.; Steck, A.K.; Palmer, J.P.; Krischer, J.P.; Geyer, S.; Xu, P.; Sosenko, J.M. β Cell dysfunction exists more than 5 years before type 1 diabetes diagnosis. J. Clin. Investig. 2018, 3, e120877. [Google Scholar] [CrossRef]
  10. Lu, X.; Xie, Q.; Pan, X.; Zhang, R.; Zhang, X.; Peng, G.; Zhang, Y.; Shen, S.; Tong, N. Type 2 diabetes mellitus in adults: Pathogenesis, prevention and therapy. Signal Transduct. Target. Ther. 2024, 9, 262. [Google Scholar] [CrossRef]
  11. Roden, M.; Shulman, G.I. The integrative biology of type 2 diabetes. Nature 2019, 576, 51–60. [Google Scholar] [CrossRef]
  12. International Diabetes Federation. IDF Diabetes Atlas: 2021, 10th ed.; International Diabetes Federation: Brussels, Belgium, 2021. [Google Scholar]
  13. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.; Mbanya, J.C.; et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res. Clin. Pract. 2022, 183, 109119. [Google Scholar] [CrossRef] [PubMed]
  14. Kautzky-Willer, A.; Harreiter, J.; Pacini, G. Sex and gender differences in risk, pathophysiology and complications of type 2 diabetes mellitus. Endocr. Rev. 2016, 37, 278–316. [Google Scholar] [CrossRef] [PubMed]
  15. Ong, K.L.; Stafford, L.K.; McLaughlin, S.A.; Boyko, E.J.; Vollset, S.E.; Smith, A.E.; Dalton, B.E.; Duprey, J.; Cruz, J.A.; Hagins, H.; et al. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the Global Burden of Disease Study 2021. Lancet 2023, 402, 203–234. [Google Scholar] [CrossRef]
  16. Bonnefond, A.; Froguel, P. Rare and common genetic events in type 2 diabetes: What should biologists know? Cell Metab. 2015, 21, 357–368. [Google Scholar] [CrossRef]
  17. McIntyre, H.D.; Catalano, P.; Zhang, C.; Desoye, G.; Mathiesen, E.R.; Damm, P. Gestational diabetes mellitus. Nat. Rev. Dis. Primers 2019, 5, 47. [Google Scholar] [CrossRef]
  18. Bonnefond, A.; Unnikrishnan, R.; Doria, A.; Vaxillaire, M.; Kulkarni, R.N.; Mohan, V.; Trischitta, V.; Froguel, P. Monogenic diabetes. Nat. Rev. Dis. Primers 2023, 9, 12. [Google Scholar] [CrossRef]
  19. Harding, J.L.; Pavkov, M.E.; Magliano, D.J.; Shaw, J.E.; Gregg, E.W. Global trends in diabetes complications: A review of current evidence. Diabetologia 2019, 62, 3–16. [Google Scholar] [CrossRef]
  20. Armstrong, D.G.; Tan, T.-W.; Boulton, A.J.; Bus, S.A. Diabetic foot ulcers: A review. JAMA 2023, 330, 62–75. [Google Scholar] [CrossRef] [PubMed]
  21. FDA-NIH. Biomarker Working Group. BEST (Biomarkers, EndpointS, and Other Tools) Resource; Food and Drug Administration, National Institutes of Health: Silver Spring, MD, USA, 2016.
  22. Califf, R.M. Biomarker definitions and their applications. Exp. Biol. Med. 2018, 243, 213–221. [Google Scholar] [CrossRef] [PubMed]
  23. Ziegler, A.G.; Hummel, M.; Schenker, M.; Bonifacio, E. Autoantibody appearance and risk for development of childhood diabetes in offspring of parents with type 1 diabetes: The 2-year analysis of the German BABYDIAB Study. Diabetes 1999, 48, 460–468. [Google Scholar] [CrossRef]
  24. Ilonen, J.; Hammais, A.; Laine, A.-P.; Lempainen, J.; Vaarala, O.; Veijola, R.; Simell, O.; Knip, M. Patterns of β-cell autoantibody appearance and genetic associations during the first years of life. Diabetes 2013, 62, 3636–3640. [Google Scholar] [CrossRef] [PubMed]
  25. Krischer, J.P.; Lynch, K.F.; Schatz, D.A.; Ilonen, J.; Lernmark, Å.; Hagopian, W.A.; Rewers, M.J.; She, J.-X.; Simell, O.G.; Toppari, J.; et al. The 6 year incidence of diabetes-associated autoantibodies in genetically at-risk children: The TEDDY study. Diabetologia 2015, 58, 980–987. [Google Scholar] [CrossRef] [PubMed]
  26. Ziegler, A.G.; Rewers, M.; Simell, O.; Simell, T.; Lempainen, J.; Steck, A.; Winkler, C.; Ilonen, J.; Veijola, R.; Knip, M.; et al. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 2013, 309, 2473–2479. [Google Scholar] [CrossRef]
  27. Erlich, H.; Valdes, A.M.; Noble, J.; Carlson, J.A.; Varney, M.; Concannon, P.; Mychaleckyj, J.C.; Todd, J.A.; Bonella, P.; Fear, A.L.; et al. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: Analysis of the type 1 diabetes genetics consortium families. Diabetes 2008, 57, 1084–1092. [Google Scholar] [CrossRef]
  28. Oliveira, S.M.R.; Rebocho, A.; Ahmadpour, E.; Nissapatorn, V.; De Lourdes Pereira, M. Type 1 diabetes mellitus: A review on advances and challenges in creating insulin producing devices. Micromachines 2023, 14, 151. [Google Scholar] [CrossRef]
  29. Sims, E.K.; Besser, R.E.; Dayan, C.; Rasmussen, C.G.; Greenbaum, C.; Griffin, K.J.; Hagopian, W.; Knip, M.; Long, A.E.; Martin, F.; et al. Screening for type 1 diabetes in the general population: A status report and perspective. Diabetes 2022, 71, 610–623. [Google Scholar] [CrossRef]
  30. American Diabetes Association. Classification and diagnosis of diabetes. Diabetes Care 2016, 38 (Suppl. S1), S8–S16. [Google Scholar]
  31. Akram, M. A focused review of the role of ketone bodies in health and disease. J. Med. Food 2013, 16, 965–967. [Google Scholar] [CrossRef] [PubMed]
  32. Saydah, S.H.; Imperatore, G.; Henkin, L.; D’agostino, R.; Divers, J.; Mayer-Davis, E.J.; Dabelea, D.; Klingensmith, G.; Pihoker, C.; Lawrence, J.M. Trends and characteristics of self-reported case presentation of diabetes diagnosis among youth from 2002 to 2010: Findings from the SEARCH for diabetes in youth study. Diabetes Care 2015, 38, e84–e85. [Google Scholar] [CrossRef][Green Version]
  33. Care, D. 2. Classification and Diagnosis of Diabetes. Diabetes Care 2015, 39, S13–S22. [Google Scholar] [CrossRef]
  34. Little, R.R.; Rohlfing, C.L. The long and winding road to optimal HbA1c measurement. Clin. Chim. Acta 2013, 418, 63–71. [Google Scholar] [CrossRef] [PubMed]
  35. Shields, B.M.; McDonald, T.J.; Oram, R.; Hill, A.; Hudson, M.; Leete, P.; Pearson, E.R.; Richardson, S.J.; Morgan, N.G.; Hattersley, A.T.; et al. C-Peptide decline in type 1 diabetes has two phases: An initial exponential fall and a subsequent stable phase. Diabetes Care 2018, 41, 1486–1492. [Google Scholar] [CrossRef]
  36. Iqbal, S.; Abu Jayyab, A.; Alrashdi, A.M.; Reverté-Villarroya, S. The predictive ability of c-peptide in distinguishing type 1 diabetes from type 2 diabetes: A systematic review and meta-analysis. Endocr. Pract. 2023, 29, 379–387. [Google Scholar] [CrossRef]
  37. American Diabetes Association. Professional Practice Committee: Standards of Medical Care in Diabetes—2022; American Diabetes Association: Arlington, VA, USA, 2022; p. S3. [Google Scholar]
  38. WHO Expert Consultation. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet 2004, 363, 157–163. [Google Scholar] [CrossRef] [PubMed]
  39. Prillaman, M. Why BMI is flawed—And how to redefine obesity. Nature 2023, 622, 232–233. [Google Scholar] [CrossRef]
  40. Jayedi, A.; Soltani, S.; Zargar, M.S.; Khan, T.A.; Shab-Bidar, S. Central fatness and risk of all cause mortality: Systematic review and dose-response meta-analysis of 72 prospective cohort studies. BMJ 2020, 370, m3324. [Google Scholar] [CrossRef]
  41. Balasubramanyam, A. Defining and Classifying New Subgroups of Diabetes. Annu. Rev. Med. 2021, 72, 63–74. [Google Scholar] [CrossRef]
  42. Xu, S.; Wu, Y.; Li, J.; Pan, X.; Zhang, X.; Liu, Y.; Zhang, F.; Tong, N. Evaluation of the value of diabetes risk scores in screening for undiagnosed diabetes and prediabetes: A community-based study in southwestern China. Postgrad. Med. 2020, 132, 737–745. [Google Scholar] [CrossRef] [PubMed]
  43. Bergman, M.; Manco, M.; Satman, I.; Chan, J.; Schmidt, M.I.; Sesti, G.; Fiorentino, T.V.; Abdul-Ghani, M.; Jagannathan, R.; Aravindakshan, P.K.T.; et al. International Diabetes Federation Position Statement on the 1-h post-load plasma glucose for the diagnosis of intermediate hyperglycaemia and type 2 diabetes. Diabetes Res. Clin. Pract. 2024, 209, 111589. [Google Scholar] [CrossRef]
  44. World Health Organization. Classification of Diabetes Mellitus; World Health Organization: Geneva, Switzerland, 2019. [Google Scholar]
  45. Rojas, J.; Bermudez, V.; Palmar, J.; Martínez, M.S.; Olivar, L.C.; Nava, M.; Tomey, D.; Rojas, M.; Salazar, J.; Garicano, C.; et al. Pancreatic beta cell death: Novel potential mechanisms in diabetes therapy. J. Diabetes Res. 2018, 2018, 9601801. [Google Scholar] [CrossRef]
  46. Cernea, S.; Dobreanu, M. Diabetes and beta cell function: From mechanisms to evaluation and clinical implications. Biochem. Medica 2013, 23, 266–280. [Google Scholar] [CrossRef]
  47. Metcalf, G.A.D. MicroRNAs: Circulating biomarkers for the early detection of imperceptible cancers via biosensor and machine-learning advances. Oncogene 2024, 43, 2135–2142. [Google Scholar] [CrossRef]
  48. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. miRNAs as biomarkers in disease: Latest findings regarding their role in diagnosis and prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, C.; Sun, C.; Zhao, Y.; Wang, Q.; Guo, J.; Ye, B.; Yu, G. Overview of MicroRNAs as diagnostic and prognostic biomarkers for high-incidence cancers in 2021. Int. J. Mol. Sci. 2022, 23, 11389. [Google Scholar] [CrossRef]
  50. Guay, C.; Regazzi, R. Circulating microRNAs as novel biomarkers for diabetes mellitus. Nat. Rev. Endocrinol. 2013, 9, 513–521. [Google Scholar] [CrossRef] [PubMed]
  51. Hardikar, A.; Joglekar, M.; Wong, W.; Kunte, P.; Kulkarni, R.; Ahmed, I.; Farr, R.J.; Pham, N.H.T.; Coles, M.; Maynard, C.L.; et al. Applicability of a microRNA-based dynamic risk score for type 1 diabetes. Res. Sq. 2024, preprint. [Google Scholar] [CrossRef]
  52. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef]
  53. Wong, W.; Farr, R.; Joglekar, M.; Januszewski, A.; Hardikar, A. Probe-based real-time PCR approaches for quantitative measurement of microRNAs. J. Vis. Exp. JoVE 2015, 98, 52586. [Google Scholar]
  54. Fyvie, M.J.; Gillespie, K.M. The importance of biomarker development for monitoring type 1 diabetes progression rate and therapeutic responsiveness. Front. Immunol. 2023, 14, 1158278. [Google Scholar] [CrossRef]
  55. Watkins, R.A.; Evans-Molina, C.; Blum, J.S.; DiMeglio, L.A. Established and emerging biomarkers for the prediction of type 1 diabetes: A systematic review. Transl. Res. 2014, 164, 110–121. [Google Scholar] [CrossRef]
  56. Joglekar, M.V.; Kaur, S.; Pociot, F.; Hardikar, A. Prediction of progression to type 1 diabetes with dynamic biomarkers and risk scores. Lancet Diabetes Endocrinol. 2024, 12, 483–492. [Google Scholar] [CrossRef]
  57. Bonifacio, E. Predicting Type 1 Diabetes Using Biomarkers. Diabetes Care 2015, 38, 989–996. [Google Scholar] [CrossRef]
  58. Yi, L.; Swensen, A.C.; Qian, W.-J. Serum biomarkers for diagnosis and prediction of type 1 diabetes. Transl. Res. 2018, 201, 13–25. [Google Scholar] [CrossRef]
  59. Brenu, E.W.; Harris, M.; Hamilton-Williams, E.E. Circulating biomarkers during progression to type 1 diabetes: A systematic review. Front. Endocrinol. 2023, 14, 1117076. [Google Scholar] [CrossRef] [PubMed]
  60. Zhu, H.; Leung, S.-W. MicroRNA biomarkers of type 2 diabetes: Evidence synthesis from meta-analyses and pathway modelling. Diabetologia 2023, 66, 288–299. [Google Scholar] [CrossRef]
  61. Alicka, M.; Major, P.; Wysocki, M.; Marycz, K. Adipose-derived mesenchymal stem cells isolated from patients with type 2 diabetes show reduced “stemness” through an altered secretome profile, impaired anti-oxidative protection, and mitochondrial dynamics deterioration. J. Clin. Med. 2019, 8, 765. [Google Scholar] [CrossRef]
  62. Lopez, Y.O.N.; Retnakaran, R.; Zinman, B.; Pratley, R.E.; Seyhan, A.A. Predicting and understanding the response to short-term intensive insulin therapy in people with early type 2 diabetes. Mol. Metab. 2019, 20, 63–78. [Google Scholar] [CrossRef]
  63. Han, L.-L.; Wang, S.-H.; Yao, M.-Y.; Zhou, H. Urinary exosomal microRNA-145-5p and microRNA-27a-3p act as noninvasive diagnostic biomarkers for diabetic kidney disease. World J. Diabetes 2024, 15, 92–104. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, J.; Cui, C.; Xu, H. Downregulation of miR-145-5p elevates retinal ganglion cell survival to delay diabetic retinopathy progress by targeting FGF5. Biosci. Biotechnol. Biochem. 2019, 83, 1655–1662. [Google Scholar] [CrossRef]
  65. Barutta, F.; Bellini, S.; Guarrera, S.; Matullo, G.; Schalkwijk, C.; Stehouwer, C.D.; Chaturvedi, N.; Soedamah-Muthu, S.S.; Durazzo, M.; Gruden, G. Association of serum MicroRNA-145-5p levels with microvascular complications of type 1 Diabetes: The EURODIAB prospective complications study. Diabetes Res. Clin. Pract. 2022, 190, 109987. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, Z.; Tao, B.; Fan, S.; Cui, S.; Pu, Y.; Qiu, L.; Xia, H.; Xu, L. Over-expression of microRNA-145 drives alterations in β-adrenergic signaling and attenuates cardiac remodeling in heart failure post myocardial infarction. Aging 2020, 12, 11603–11622. [Google Scholar] [CrossRef]
  67. Liang, L.; Stone, R.C.; Stojadinovic, O.; Ramirez, H.; Pastar, I.; Maione, A.G.; Smith, A.; Yanez, V.; Veves, A.; Kirsner, R.S.; et al. Integrative analysis of miRNA and mRNA paired expression profiling of primary fibroblast derived from diabetic foot ulcers reveals multiple impaired cellular functions. Wound Repair Regen. 2016, 24, 943–953. [Google Scholar] [CrossRef]
  68. Wang, C.; Huang, L.; Li, J.; Liu, D.; Wu, B. MicroRNA miR-145-5p inhibits cutaneous wound healing by targeting PDGFD in diabetic foot ulcer. Biochem. Genet. 2024, 62, 2437–2454. [Google Scholar] [CrossRef]
  69. Hromadnikova, I.; Kotlabova, K.; Krofta, L. Cardiovascular disease-associated MicroRNAs as novel biomarkers of first-trimester screening for gestational diabetes mellitus in the absence of other pregnancy-related complications. Int. J. Mol. Sci. 2022, 23, 10635. [Google Scholar] [CrossRef] [PubMed]
  70. Åkerman, L.; Casas, R.; Ludvigsson, J.; Tavira, B.; Skoglund, C.; van Wijnen, A. Serum miRNA levels are related to glucose homeostasis and islet autoantibodies in children with high risk for type 1 diabetes. PLoS ONE 2018, 13, e0191067. [Google Scholar] [CrossRef]
  71. Weale, C.J.; Matshazi, D.M.; Davids, S.F.G.; Raghubeer, S.; Erasmus, R.T.; Kengne, A.P.; Davison, G.M.; Matsha, T.E. MicroRNAs-1299, -126-3p and -30e-3p as potential diagnostic biomarkers for prediabetes. Diagnostics 2021, 11, 949. [Google Scholar] [CrossRef]
  72. Weale, C.J.; Matshazi, D.M.; Davids, S.F.G.; Raghubeer, S.; Erasmus, R.T.; Kengne, A.P.; Davison, G.M.; Matsha, T.E. Expression profiles of circulating microRNAs in South African type 2 diabetic individuals on treatment. Front. Genet. 2021, 12, 702410. [Google Scholar] [CrossRef] [PubMed]
  73. Zeng, Q.; Liu, J. Silencing circ_0001879 inhibits the proliferation and migration of human retinal microvascular endothelial cells under high-glucose conditions via modulating miR-30-3p. Gene 2020, 760, 144992. [Google Scholar] [CrossRef]
  74. Samidurai, A.; Olex, A.L.; Ockaili, R.; Kraskauskas, D.; Roh, S.K.; Kukreja, R.C.; Das, A. Integrated Analysis of lncRNA–miRNA–mRNA Regulatory Network in Rapamycin-Induced Cardioprotection against Ischemia/Reperfusion Injury in Diabetic Rabbits. Cells 2023, 12, 2820. [Google Scholar] [CrossRef] [PubMed]
  75. Desjarlais, M.; Rivera, J.C.; Lahaie, I.; Cagnone, G.; Wirt, M.; Omri, S.; Chemtob, S.; Chen, J. MicroRNA expression profile in retina and choroid in oxygen-induced retinopathy model. PLoS ONE 2019, 14, e0218282. [Google Scholar] [CrossRef]
  76. Motshwari, D.D.; George, C.; Matshazi, D.M.; Weale, C.J.; Davids, S.F.G.; Zemlin, A.E.; Erasmus, R.T.; Kengne, A.P.; Matsha, T.E. Expression of whole blood miR-126-3p, -30a-5p, -1299, -182-5p and -30e-3p in chronic kidney disease in a South African community-based sample. Sci. Rep. 2022, 12, 4107. [Google Scholar] [CrossRef]
  77. Grieco, G.E.; Besharat, Z.M.; Licata, G.; Fignani, D.; Brusco, N.; Nigi, L.; Formichi, C.; Po, A.; Sabato, C.; Dardano, A.; et al. Circulating microRNAs as clinically useful biomarkers for Type 2 Diabetes Mellitus: miRNomics from bench to bedside. Transl. Res. 2022, 247, 137–157. [Google Scholar] [CrossRef]
  78. Stępień, E.Ł.; Durak-Kozica, M.; Kamińska, A.; Targosz-Korecka, M.; Libera, M.; Tylko, G.; Opalińska, A.; Kapusta, M.; Solnica, B.; Georgescu, A.; et al. Circulating ectosomes: Determination of angiogenic microRNAs in type 2 diabetes. Theranostics 2018, 8, 3874–3890. [Google Scholar] [CrossRef] [PubMed]
  79. Wang, J.; Duan, L.; Guo, T.; Gao, Y.; Tian, L.; Liu, J.; Wang, S.; Yang, J. Downregulation of miR-30c promotes renal fibrosis by target CTGF in diabetic nephropathy. J. Diabetes Its Complicat. 2016, 30, 406–414. [Google Scholar] [CrossRef] [PubMed]
  80. Ghai, V.; Wu, X.; Bheda-Malge, A.; Argyropoulos, C.P.; Bernardo, J.F.; Orchard, T.; Galas, D.; Wang, K. Genome-wide profiling of urinary extracellular vesicle microRNAs associated with diabetic nephropathy in type 1 diabetes. Kidney Int. Rep. 2018, 3, 555–572. [Google Scholar] [CrossRef] [PubMed]
  81. Assmann, T.S.; Recamonde-Mendoza, M.; De Souza, B.M.; Crispim, D. MicroRNA expression profiles and type 1 diabetes mellitus: Systematic review and bioinformatic analysis. Endocr. Connect. 2017, 6, 773–790. [Google Scholar] [CrossRef]
  82. Nielsen, L.B.; Wang, C.; Sørensen, K.; Bang-Berthelsen, C.H.; Hansen, L.; Andersen, M.-L.M.; Hougaard, P.; Juul, A.; Zhang, C.-Y.; Pociot, F.; et al. Circulating levels of MicroRNA from children with newly diagnosed type 1 diabetes and healthy controls: Evidence that miR-25 associates to residual beta-cell function and glycaemic control during disease progression. Exp. Diabetes Res. 2012, 2012, 896362. [Google Scholar] [CrossRef]
  83. Demirsoy, H.; Ertural, D.Y.; Balci, Ş.; Çınkır, Ü.; Sezer, K.; Tamer, L.; Aras, N. Profiles of circulating miRNAs following metformin treatment in patients with type 2 diabetes. J. Med. Biochem. 2018, 37, 499. [Google Scholar] [CrossRef]
  84. Guay, C.; Kruit, J.K.; Rome, S.; Menoud, V.; Mulder, N.L.; Jurdzinski, A.; Mancarella, F.; Sebastiani, G.; Donda, A.; Gonzalez, B.J.; et al. Lymphocyte-derived exosomal MicroRNAs promote pancreatic β cell death and may contribute to type 1 diabetes development. Cell Metab. 2019, 29, 348–361. [Google Scholar] [CrossRef]
  85. Mostahfezian, M.; Azhir, Z.; Dehghanian, F.; Hojati, Z. Expression Pattern of microRNAs, miR-21, miR-155 and miR-338 in patients with type 1 diabetes. Arch. Med Res. 2019, 50, 79–85. [Google Scholar] [CrossRef]
  86. Catanzaro, G.; Conte, F.; Trocchianesi, S.; Splendiani, E.; Bimonte, V.M.; Mocini, E.; Filardi, T.; Po, A.; Besharat, Z.M.; Gentile, M.C.; et al. Network analysis identifies circulating miR-155 as predictive biomarker of type 2 diabetes mellitus development in obese patients: A pilot study. Sci. Rep. 2023, 13, 19496. [Google Scholar] [CrossRef]
  87. Nemecz, M.; Stefan, D.S.; Comarița, I.K.; Constantin, A.; Tanko, G.; Guja, C.; Georgescu, A. Microvesicle-associated and circulating microRNAs in diabetic dyslipidemia: miR-218, miR-132, miR-143, and miR-21, miR-122, miR-155 have biomarker potential. Cardiovasc. Diabetol. 2023, 22, 260. [Google Scholar] [CrossRef] [PubMed]
  88. Xu, M.; Li, Y.; Tang, Y.; Zhao, X.; Xie, D.; Chen, M. Increased expression of miR-155 in peripheral blood and wound margin tissue of type 2 diabetes mellitus patients associated with diabetic foot ulcer. Diabetes Metab. Syndr. Obes. Targets Ther. 2022, 15, 3415–3428. [Google Scholar] [CrossRef]
  89. Gholaminejad, A.; Tehrani, H.A.; Fesharaki, M.G. Identification of candidate microRNA biomarkers in diabetic nephropathy: A meta-analysis of profiling studies. J. Nephrol. 2018, 31, 813–831. [Google Scholar] [CrossRef]
  90. Santovito, D.; Toto, L.; De Nardis, V.; Marcantonio, P.; D’aLoisio, R.; Mastropasqua, A.; De Cesare, D.; Bucci, M.; Paganelli, C.; Natarelli, L.; et al. Plasma microRNA signature associated with retinopathy in patients with type 2 diabetes. Sci. Rep. 2021, 11, 4136. [Google Scholar] [CrossRef]
  91. Bi, Y.; Yang, S.; Zhang, Z. 127-OR: Identification of Exosomal miRNA as a Novel Causal Biomarker for Cognitive Impairment in Type 2 Diabetes Mellitus—Triangulating Evidence from Mendelian Randomization Analyses and Multicenter Cohort. Diabetes 2024, 73 (Suppl. S1), 127-OR. [Google Scholar] [CrossRef]
  92. Lin, G.; Liu, X.; Hasan, M. Key extracellular proteins and TF-miRNA co-regulatory network in diabetic foot ulcer: Bioinformatics and experimental insights. PLoS ONE 2024, 19, e0307205. [Google Scholar] [CrossRef] [PubMed]
  93. Pheiffer, C.; Dias, S.; Rheeder, P.; Adam, S. Decreased expression of circulating miR-20a-5p in South African women with gestational diabetes mellitus. Mol. Diagn. Ther. 2018, 22, 345–352. [Google Scholar] [CrossRef]
  94. Jin, Z.-Q. MicroRNA targets and biomarker validation for diabetes-associated cardiac fibrosis. Pharmacol. Res. 2021, 174, 105941. [Google Scholar] [CrossRef]
  95. Liu, X.; Guo, B.; Zhang, W.; Ma, B.; Li, Y. MiR-20a-5p overexpression prevented diabetic cardiomyopathy via inhibition of cardiomyocyte apoptosis, hypertrophy, fibrosis and JNK/NF-κB signalling pathway. J. Biochem. 2021, 170, 349–362. [Google Scholar] [CrossRef] [PubMed]
  96. Ye, D.; Zhang, T.; Lou, G.; Xu, W.; Dong, F.; Chen, G.; Liu, Y. Plasma miR-17, miR-20a, miR-20b and miR-122 as potential biomarkers for diagnosis of NAFLD in type 2 diabetes mellitus patients. Life Sci. 2018, 208, 201–207. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, H.; Niu, Q.; Liang, K.; Li, X.; Jiang, J.; Bian, C. Effect of LncPVT1/miR-20a-5p on lipid metabolism and insulin resistance in NAFLD. Diabetes Metab. Syndr. Obes. Targets Ther. 2021, 14, 4599–4608. [Google Scholar] [CrossRef]
  98. Sebastiani, G.; Grieco, F.A.; Spagnuolo, I.; Galleri, L.; Cataldo, D.; Dotta, F. Increased expression of microRNA miR-326 in type 1 diabetic patients with ongoing islet autoimmunity. Diabetes/Metab. Res. Rev. 2011, 27, 862–866. [Google Scholar] [CrossRef]
  99. Erener, S.; Marwaha, A.; Tan, R.; Panagiotopoulos, C.; Kieffer, T.J. Profiling of circulating microRNAs in children with recent onset of type 1 diabetes. J. Clin. Investig. 2017, 2, e89656. [Google Scholar] [CrossRef]
  100. Blanco, J.A.; Lambert, C.; Fernandez-Sanjurjo, M.; Morales-Sanchez, P.; Pujante, P.; Pinto-Hernández, P.; Iglesias-Gutiérrez, E.; Torre, E.M.; Delgado, E. miR-24-3p and body mass index as type 2 diabetes risk factors in spanish women 15 years after gestational diabetes mellitus diagnosis. Int. J. Mol. Sci. 2023, 24, 1152. [Google Scholar] [CrossRef]
  101. Guo, J.; Zhou, P.; Pan, M.; Liu, Z.; An, G.; Han, J.; Dai, F.; Du, L.; Jin, X. Relationship between elevated microRNAs and growth factors levels in the vitreous of patients with proliferative diabetic retinopathy. J. Diabetes Its Complicat. 2021, 35, 108021. [Google Scholar] [CrossRef] [PubMed]
  102. Guo, J.; Zhou, P.; Liu, Z.; Dai, F.; Pan, M.; An, G.; Han, J.; Du, L.; Jin, X. The aflibercept-induced MicroRNA profile in the vitreous of proliferative diabetic retinopathy patients detected by next-generation sequencing. Front. Pharmacol. 2021, 12, 781276. [Google Scholar] [CrossRef] [PubMed]
  103. Meng, F.; Shen, F.; Ling, H.; Jin, P.; Zhou, D.; Li, Q. CircARHGAP12 triggers mesenchymal stromal cell autophagy to facilitate its effect on repairing diabetic wounds by sponging miR-301b-3p/ATG16L1 and miR-301b-3p/ULK2. J. Investig. Dermatol. 2022, 142, 1976–1989.e4. [Google Scholar] [CrossRef]
  104. Sánchez-Ceinos, J.; Rangel-Zuñiga, O.A.; Clemente-Postigo, M.; Podadera-Herreros, A.; Camargo, A.; Alcalá-Diaz, J.F.; Guzmán-Ruiz, R.; López-Miranda, J.; Malagón, M.M. miR-223-3p as a potential biomarker and player for adipose tissue dysfunction preceding type 2 diabetes onset. Mol. Ther. Nucleic Acids 2021, 23, 1035–1052. [Google Scholar] [CrossRef]
  105. Da’aS, S.I.; Ahmed, I.; Hasan, W.H.; Abdelrahman, D.A.; Aliyev, E.; Nisar, S.; Bhat, A.A.; Joglekar, M.V.; Hardikar, A.A.; Fakhro, K.A.; et al. The link between glycemic control measures and eye microvascular complications in a clinical cohort of type 2 diabetes with microRNA-223-3p signature. J. Transl. Med. 2023, 21, 171. [Google Scholar] [CrossRef]
  106. Abdeltawab, A.; Zaki, M.; Abdeldayem, Y.; Mohamed, A.; Zaied, S.M. Circulating micro RNA-223 and angiopoietin-like protein 8 as biomarkers of gestational diabetes mellitus. Br. J. Biomed. Sci. 2021, 78, 12–17. [Google Scholar] [CrossRef]
  107. Zang, J.; Maxwell, A.P.; Simpson, D.A.; McKay, G.J. Differential expression of urinary exosomal MicroRNAs miR-21-5p and miR-30b-5p in individuals with diabetic kidney disease. Sci. Rep. 2019, 9, 10900. [Google Scholar] [CrossRef]
  108. Pordzik, J.; Jakubik, D.; Jarosz-Popek, J.; Wicik, Z.; Eyileten, C.; De Rosa, S.; Indolfi, C.; Siller-Matula, J.M.; Czajka, P.; Postula, M. Significance of circulating microRNAs in diabetes mellitus type 2 and platelet reactivity: Bioinformatic analysis and review. Cardiovasc. Diabetol. 2019, 18, 113. [Google Scholar] [CrossRef] [PubMed]
  109. Weale, C.J.; Matshazi, D.M.; Davids, S.F.; Raghubeer, S.; Erasmus, R.T.; Kengne, A.P.; Davison, G.M.; Matsha, T.E. Circulating miR-30a-5p and miR-182-5p in prediabetes and screen-detected diabetes mellitus. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 5037–5047. [Google Scholar] [CrossRef]
  110. Prabu, P.; Rome, S.; Sathishkumar, C.; Gastebois, C.; Meugnier, E.; Mohan, V.; Balasubramanyam, M. MicroRNAs from urinary extracellular vesicles are non-invasive early biomarkers of diabetic nephropathy in type 2 diabetes patients with the ‘Asian Indian phenotype’. Diabetes Metab. 2019, 45, 276–285. [Google Scholar] [CrossRef] [PubMed]
  111. Guo, B.; Xu, X.; Chi, X.; Wang, M. Relationship of lncRNA FTX and miR-186-5p levels with diabetic peripheral neuropathy in type 2 diabetes and its bioinformatics analysis. Ir. J. Med Sci. 2024, 193, 2293–2299. [Google Scholar] [CrossRef]
  112. Jiang, J.; Mo, H.; Liu, C.; Wu, B.; Wu, Z.; Li, X.; Li, T.; He, S.; Li, S.; You, Q.; et al. Inhibition of miR-186-5p contributes to high glucose-induced injury in AC16 cardiomyocytes. Exp. Ther. Med. 2018, 15, 627–632. [Google Scholar] [CrossRef]
  113. Wang, K.-J.; Zhao, X.; Liu, Y.-Z.; Zeng, Q.-T.; Mao, X.-B.; Li, S.-N.; Zhang, M.; Jiang, C.; Zhou, Y.; Qian, C.; et al. Circulating MiR-19b-3p, MiR-134-5p and MiR-186-5p are promising novel biomarkers for early diagnosis of acute myocardial infarction. Cell. Physiol. Biochem. 2016, 38, 1015–1029. [Google Scholar] [CrossRef]
  114. Yuan, Y.; Guan, Y.; Shao, C.; Wang, H.; Zhang, S. Circ_0001953 contribute to retinal vascular endothelial cell injury induced by high glucose through regulating miR 186. Trop. J. Pharm. Res. 2023, 22, 563–569. [Google Scholar] [CrossRef]
  115. Olivieri, F.; Bonafè, M.; Spazzafumo, L.; Gobbi, M.; Prattichizzo, F.; Recchioni, R.; Marcheselli, F.; La Sala, L.; Galeazzi, R.; Rippo, M.R.; et al. Age- and glycemia-related miR-126-3p levels in plasma and endothelial cells. Aging 2014, 6, 771–786. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, T.; Lv, C.; Li, L.; Chen, S.; Liu, S.; Wang, C.; Su, B. Plasma miR-126 is a potential biomarker for early prediction of type 2 diabetes mellitus in susceptible individuals. BioMed Res. Int. 2013, 2013, 761617. [Google Scholar] [CrossRef]
  117. Ghaneh, T.; Zeinali, F.; Babini, H.; Astaraki, S.; Hassan-Zadeh, V. An increase in the expression of circulating miR30d-5p and miR126-3p is associated with intermediate hyperglycaemia in Iranian population. Arch. Physiol. Biochem. 2023, 129, 489–496. [Google Scholar] [CrossRef] [PubMed]
  118. Qin, L.-L.; An, M.-X.; Liu, Y.-L.; Xu, H.-C.; Lu, Z.-Q. MicroRNA-126: A promising novel biomarker in peripheral blood for diabetic retinopathy. Int. J. Ophthalmol. 2017, 10, 530–534. [Google Scholar] [CrossRef] [PubMed]
  119. Barutta, F.; Bruno, G.; Matullo, G.; Chaturvedi, N.; Grimaldi, S.; Schalkwijk, C.; Stehouwer, C.D.; Fuller, J.H.; Gruden, G. MicroRNA-126 and micro-/macrovascular complications of type 1 diabetes in the EURODIAB Prospective Complications Study. Acta Diabetol. 2017, 54, 133–139. [Google Scholar] [CrossRef] [PubMed]
  120. Sebastiani, G.; Grieco, G.E.; Bruttini, M.; Auddino, S.; Mori, A.; Toniolli, M.; Fignani, D.; Licata, G.; Aiello, E.; Nigi, L.; et al. A set of circulating microRNAs belonging to the 14q32 chromosome locus identifies two subgroups of individuals with recent-onset type 1 diabetes. Cell Rep. Med. 2024, 5, 101591. [Google Scholar] [CrossRef]
  121. Strycharz, J.; Wróblewski, A.; Zieleniak, A.; Świderska, E.; Matyjas, T.; Rucińska, M.; Pomorski, L.; Czarny, P.; Szemraj, J.; Drzewoski, J.; et al. Visceral adipose tissue of prediabetic and diabetic females shares a set of similarly upregulated microRNAs functionally annotated to inflammation, oxidative stress and insulin signaling. Antioxidants 2021, 10, 101. [Google Scholar] [CrossRef]
  122. Lovis, P.; Roggli, E.; Laybutt, D.R.; Gattesco, S.; Yang, J.-Y.; Widmann, C.; Abderrahmani, A.; Regazzi, R. Alterations in MicroRNA expression contribute to fatty acid–induced pancreatic β-cell dysfunction. Diabetes 2008, 57, 2728–2736. [Google Scholar] [CrossRef]
  123. Rong, Y.; Bao, W.; Shan, Z.; Liu, J.; Yu, X.; Xia, S.; Gao, H.; Wang, X.; Yao, P.; Hu, F.B.; et al. Increased MicroRNA-146a levels in plasma of patients with newly diagnosed type 2 diabetes mellitus. PLoS ONE 2013, 8, e73272. [Google Scholar] [CrossRef]
  124. Kong, L.; Zhu, J.; Han, W.; Jiang, X.; Xu, M.; Zhao, Y.; Dong, Q.; Pang, Z.; Guan, Q.; Gao, L.; et al. Significance of serum microRNAs in pre-diabetes and newly diagnosed type 2 diabetes: A clinical study. Acta Diabetol. 2011, 48, 61–69. [Google Scholar] [CrossRef]
  125. Assmann, T.S.; Recamonde-Mendoza, M.; Puñales, M.; Tschiedel, B.; Canani, L.H.; Crispim, D. MicroRNA expression profile in plasma from type 1 diabetic patients: Case-control study and bioinformatic analysis. Diabetes Res. Clin. Pract. 2018, 141, 35–46. [Google Scholar] [CrossRef]
  126. Xue, L.; Xiong, C.; Li, J.; Ren, Y.; Zhang, L.; Jiao, K.; Chen, C.; Ding, P. miR-200-3p suppresses cell proliferation and reduces apoptosis in diabetic retinopathy via blocking the TGF-β2/Smad pathway. Biosci. Rep. 2020, 40, BSR20201545. [Google Scholar] [CrossRef] [PubMed]
  127. Civantos, E.; Bosch, E.; Bustillo, E.R.; Zhenyukh, O.; Egido, J.; Lorenzo, O.; Mas, S. Sitagliptin ameliorates oxidative stress in experimental diabetic nephropathy by diminishing the miR-200a/Keap-1/Nrf2 antioxidant pathway. Diabetes Metab. Syndr. Obes. Targets Ther. 2017, 10, 207–222. [Google Scholar] [CrossRef]
  128. Gholaminejad, A.; Tehrani, H.A.; Fesharaki, M.G. Identification of candidate microRNA biomarkers in renal fibrosis: A meta-analysis of profiling studies. Biomarkers 2018, 23, 713–724. [Google Scholar] [CrossRef] [PubMed]
  129. You, P.; Chen, H.; Han, W.; Deng, J. miR-200a-3p overexpression alleviates diabetic cardiomyopathy injury in mice by regulating autophagy through the FOXO3/Mst1/Sirt3/AMPK axis. PeerJ 2023, 11, e15840. [Google Scholar] [CrossRef]
  130. Kim, K.W.; Ho, A.; Alshabee-Akil, A.; Hardikar, A.A.; Kay, T.W.; Rawlinson, W.D.; Craig, M.E. Coxsackievirus B5 infection induces dysregulation of microRNAs predicted to target known type 1 diabetes risk genes in human pancreatic islets. Diabetes 2016, 65, 996–1003. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, X.; Zhao, Z.; Chen, D.; Zhang, Z.; Lin, X.; Shen, Z.; Lin, Q.; Fan, K.; Wang, Q.; Zhang, W.; et al. SIRT1 and miR-34a-5p expression in PBMCs as potential biomarkers for patients with type 2 diabetes with cognitive impairments. J. Clin. Endocrinol. Metab. 2024, 109, 815–826. [Google Scholar] [CrossRef]
  132. Milcu, A.I.; Anghel, F.M.; Romanescu, M.; Chis, A.R.; Anghel, A.; Boruga, O. Plasma miR-19b, miR-34a, and miR-146a expression in patients with type 2 diabetes mellitus and cataract: A pilot study. Biomol. Biomed. 2024, 24, 537–544. [Google Scholar] [CrossRef]
  133. Klöting, N.; Berthold, S.; Kovacs, P.; Schön, M.R.; Fasshauer, M.; Ruschke, K.; Stumvoll, M.; Blüher, M. MicroRNA expression in human omental and subcutaneous adipose tissue. PLoS ONE 2009, 4, e4699. [Google Scholar] [CrossRef]
  134. Liang, X.; Xu, W. miR-181a-5p regulates the proliferation and apoptosis of glomerular mesangial cells by targeting KLF6. Exp. Ther. Med. 2020, 20, 1121–1128. [Google Scholar] [CrossRef]
  135. Li, H.; Zhang, P.; Li, F.; Yuan, G.; Wang, X.; Zhang, A.; Li, F. Plasma miR-22-5p, miR-132-5p, and miR-150-3p Are associated with acute myocardial infarction. BioMed Res. Int. 2019, 2019, 5012648. [Google Scholar] [CrossRef]
  136. Giannella, A.; Castelblanco, E.; Zambon, C.F.; Basso, D.; Hernandez, M.; Ortega, E.; Alonso, N.; Mauricio, D.; Avogaro, A.; Ceolotto, G.; et al. Circulating small noncoding RNA profiling as a potential biomarker of atherosclerotic plaque composition in type 1 diabetes. Diabetes Care 2023, 46, 551–560. [Google Scholar] [CrossRef]
  137. Yu, G.; Tam, H.C.H.; Huang, C.; Shi, M.; Lim, C.K.P.; Chan, J.C.N.; Ma, R.C.W. Lessons and Applications of Omics Research in Diabetes Epidemiology. Curr. Diabetes Rep. 2024, 24, 27–44. [Google Scholar] [CrossRef]
  138. Laakso, M. Biomarkers for type 2 diabetes. Mol. Metab. 2019, 27, S139–S146. [Google Scholar] [CrossRef]
  139. Mandal, S. New molecular biomarkers in precise diagnosis and therapy of Type 2 diabetes. Health Technol. 2019, 10, 601–608. [Google Scholar] [CrossRef]
  140. Ortiz-Martínez, M.; González-González, M.; Martagón, A.J.; Hlavinka, V.; Willson, R.C.; Rito-Palomares, M. Recent developments in biomarkers for diagnosis and screening of type 2 diabetes mellitus. Curr. Diabetes Rep. 2022, 22, 95–115. [Google Scholar] [CrossRef]
  141. Dorcely, B.; Katz, K.; Jagannathan, R.; Chiang, S.S.; Oluwadare, B.; Goldberg, I.J.; Bergman, M. Novel biomarkers for prediabetes, diabetes, and associated complications. Diabetes Metab. Syndr. Obes. Targets Ther. 2017, 10, 345–361. [Google Scholar] [CrossRef]
  142. Bhatia, P.; Raina, S.; Chugh, J.; Sharma, S. miRNAs: Early prognostic biomarkers for Type 2 diabetes mellitus? Biomark. Med. 2015, 9, 1025–1040. [Google Scholar] [CrossRef]
  143. Le, T.N.; Bright, R.; Truong, V.; Li, J.; Juneja, R.; Vasilev, K. Key biomarkers in type 2 diabetes patients: A systematic review. Diabetes Obes. Metab. 2025, 27, 7–22. [Google Scholar] [CrossRef] [PubMed]
  144. Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.B.; Ostolaza, H.; Martín, C. Pathophysiology of type 2 diabetes mellitus. Int. J. Mol. Sci. 2020, 21, 6275. [Google Scholar] [CrossRef]
  145. Jiménez-Sánchez, C.; Mezza, T.; Sinturel, F.; Li, L.; Di Giuseppe, G.; Quero, G.; Jornayvaz, F.R.; Guessous, I.; Dibner, C.; Schrauwen, P.; et al. Circulating 1,5-anhydroglucitol as a biomarker of ß-cell mass independent of a diabetes phenotype in human subjects. J. Clin. Endocrinol. Metab. 2022, 107, 2833–2843. [Google Scholar] [CrossRef]
  146. Selvin, E.; Rawlings, A.M.; Grams, M.; Klein, R.; Sharrett, A.R.; Steffes, M.; Coresh, J. Prognostic utility of fructosamine and glycated albumin for incident diabetes and microvascular complications. Lancet. Diabetes Endocrinol. 2014, 2, 279. [Google Scholar] [CrossRef]
  147. Wahid, S.T.; Sultan, J.; Handley, G.; Saeed, B.O.; Weaver, J.U.; Robinson, A.C.J. Serum fructosamine as a marker of 5-year risk of developing diabetes mellitus in patients exhibiting stress hyperglycaemia. Diabet. Med. 2002, 19, 543–548. [Google Scholar] [CrossRef]
  148. Koga, M. Glycated albumin; clinical usefulness. Clin. Chim. Acta 2014, 433, 96–104. [Google Scholar] [CrossRef]
  149. Weyer, C.; Hanson, R.L.; Tataranni, P.; Bogardus, C.; Pratley, R. A high fasting plasma insulin concentration predicts type 2 diabetes independent of insulin resistance: Evidence for a pathogenic role of relative hyperinsulinemia. Diabetes 2000, 49, 2094–2101. [Google Scholar] [CrossRef]
  150. Sokooti, S.; Kieneker, L.M.; de Borst, M.H.; Kobold, A.M.; Kootstra-Ros, J.E.; Gloerich, J.; van Gool, A.J.; Heerspink, H.J.L.; Gansevoort, R.T.; Dullaart, R.P.; et al. Plasma C-peptide and risk of developing type 2 diabetes in the general population. J. Clin. Med. 2020, 9, 3001. [Google Scholar] [CrossRef]
  151. González-González, J.G.; Violante-Cumpa, J.R.; Zambrano-Lucio, M.; Burciaga-Jimenez, E.; Castillo-Morales, P.L.; Garcia-Campa, M.; Solis, R.C.; González-Colmenero, A.D.; Rodríguez-Gutiérrez, R. HOMA-IR as a predictor of health outcomes in patients with metabolic risk factors: A systematic review and meta-analysis. High Blood Press. Cardiovasc. Prev. 2022, 29, 547–564. [Google Scholar] [CrossRef]
  152. Wareham, N.J.; Byrne, C.D.; Williams, R.; Day, N.; Hales, C.N. Fasting proinsulin concentrations predict the development of type 2 diabetes. Diabetes Care 1999, 22, 262–270. [Google Scholar] [CrossRef]
  153. Vangipurapu, J.; Stančáková, A.; Kuulasmaa, T.; Kuusisto, J.; Laakso, M.; Kulkarni, R. Both fasting and glucose-stimulated proinsulin levels predict hyperglycemia and incident type 2 diabetes: A population-based study of 9,396 finnish men. PLoS ONE 2015, 10, e0124028. [Google Scholar] [CrossRef] [PubMed]
  154. Lynch, C.J.; Adams, S.H. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat. Rev. Endocrinol. 2014, 10, 723–736. [Google Scholar] [CrossRef]
  155. Sun, L.; Liang, L.; Gao, X.; Zhang, H.; Yao, P.; Hu, Y.; Ma, Y.; Wang, F.; Jin, Q.; Li, H.; et al. Early prediction of developing type 2 diabetes by plasma acylcarnitines: A population-based study. Diabetes Care 2016, 39, 1563–1570. [Google Scholar] [CrossRef]
  156. Jäger, S.; Cuadrat, R.; Wittenbecher, C.; Floegel, A.; Hoffmann, P.; Prehn, C.; Adamski, J.; Pischon, T.; Schulze, M.B. Mendelian randomization study on amino acid metabolism suggests tyrosine as causal trait for type 2 diabetes. Nutrients 2020, 12, 3890. [Google Scholar] [CrossRef] [PubMed]
  157. Zhou, Q.; Sun, W.-W.; Chen, J.-C.; Zhang, H.-L.; Liu, J.; Lin, Y.; Lin, P.-C.; Wu, B.-X.; An, Y.-P.; Huang, L.; et al. Phenylalanine impairs insulin signaling and inhibits glucose uptake through modification of IRβ. Nat. Commun. 2022, 13, 4291. [Google Scholar] [CrossRef]
  158. Wittemans, L.B.L.; Lotta, L.A.; Oliver-Williams, C.; Stewart, I.D.; Surendran, P.; Karthikeyan, S.; Day, F.R.; Koulman, A.; Imamura, F.; Zeng, L.; et al. Assessing the causal association of glycine with risk of cardio-metabolic diseases. Nat. Commun. 2019, 10, 1060. [Google Scholar] [CrossRef]
  159. Chauhan, G.; Tabassum, R.; Mahajan, A.; Dwivedi, O.P.; Mahendran, Y.; Kaur, I.; Nigam, S.; Dubey, H.; Varma, B.; Madhu, S.V.; et al. Common variants of FTO and the risk of obesity and type 2 diabetes in Indians. J. Hum. Genet. 2011, 56, 720–726. [Google Scholar] [CrossRef]
  160. Sarhangi, N.; Sharifi, F.; Hashemian, L.; Doabsari, M.H.; Heshmatzad, K.; Rahbaran, M.; Jamaldini, S.H.; Meybodi, H.R.A.; Hasanzad, M. PPARG (Pro12Ala) genetic variant and risk of T2DM: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 12764. [Google Scholar] [CrossRef]
  161. Moazzam-Jazi, M.; Najd-Hassan-Bonab, L.; Masjoudi, S.; Tohidi, M.; Hedayati, M.; Azizi, F.; Daneshpour, M.S. Risk of type 2 diabetes and KCNJ11 gene polymorphisms: A nested case–control study and meta-analysis. Sci. Rep. 2022, 12, 20709. [Google Scholar] [CrossRef]
  162. del Bosque-Plata, L.; Martínez-Martínez, E.; Espinoza-Camacho, M.Á.; Gragnoli, C. The role of TCF7L2 in type 2 diabetes. Diabetes 2021, 70, 1220–1228. [Google Scholar] [CrossRef]
  163. Flannick, J.; Thorleifsson, G.; Beer, N.L.; Jacobs, S.B.R.; Grarup, N.; Burtt, N.P.; Mahajan, A.; Fuchsberger, C.; Atzmon, G.; Benediktsson, R.; et al. Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nat. Genet. 2014, 46, 357–363. [Google Scholar] [CrossRef]
  164. Wei, F.; Cai, C.; Feng, S.; Lv, J.; Li, S.; Chang, B.; Zhang, H.; Shi, W.; Han, H.; Ling, C.; et al. TOX and CDKN2A/B gene polymorphisms are associated with type 2 diabetes in Han Chinese. Sci. Rep. 2015, 5, 11900. [Google Scholar] [CrossRef] [PubMed]
  165. van Vliet-Ostaptchouk, J.V.; Onland-Moret, N.C.; van Haeften, T.W.; Franke, L.; Elbers, C.C.; Shiri-Sverdlov, R.; van der Schouw, Y.T.; Hofker, M.H.; Wijmenga, C. HHEX gene polymorphisms are associated with type 2 diabetes in the Dutch Breda cohort. Eur. J. Hum. Genet. 2008, 16, 652–656. [Google Scholar] [CrossRef]
  166. Huang, Q.; Yin, J.-Y.; Dai, X.-P.; Pei, Q.; Dong, M.; Zhou, Z.-G.; Huang, X.; Yu, M.; Zhou, H.-H.; Liu, Z.-Q. IGF2BP2 variations influence repaglinide response and risk of type 2 diabetes in Chinese population. Acta Pharmacol. Sin. 2010, 31, 709–717. [Google Scholar] [CrossRef] [PubMed]
  167. Rodríguez-Morán, M.; Guerrero-Romero, F. increased levels of c-reactive protein in noncontrolled type II diabetic subjects. J. Diabetes Its Complicat. 1999, 13, 211–215. [Google Scholar] [CrossRef] [PubMed]
  168. Pradhan, A.D.; Manson, J.E.; Rifai, N.; Buring, J.E.; Ridker, P.M. C-reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 2001, 286, 327–334. [Google Scholar] [CrossRef]
  169. Fève, B.; Bastard, J.-P. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2009, 5, 305–311. [Google Scholar] [CrossRef]
  170. Moller, D. Potential Role of TNF-α in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol. Metab. 2000, 11, 212–217. [Google Scholar] [CrossRef]
  171. Masters, S.L.; Dunne, A.; Subramanian, S.L.; Hull, R.L.; Tannahill, G.M.; A Sharp, F.; Becker, C.; Franchi, L.; Yoshihara, E.; Chen, Z.; et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1β in type 2 diabetes. Nat. Immunol. 2010, 11, 897–904. [Google Scholar] [CrossRef]
  172. Panee, J. Monocyte Chemoattractant Protein 1 (MCP-1) in obesity and diabetes. Cytokine 2012, 60, 1–12. [Google Scholar] [CrossRef] [PubMed]
  173. Yoon, H.S.; Cho, C.H.; Yun, M.S.; Jang, S.J.; You, H.J.; Kim, J.-H.; Han, D.; Cha, K.H.; Moon, S.H.; Lee, K.; et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 2021, 6, 563–573. [Google Scholar] [CrossRef]
  174. Graessler, J.; Qin, Y.; Zhong, H.; Zhang, J.; Licinio, J.; Wong, M.-L.; Xu, A.; Chavakis, T.; Bornstein, A.B.; Ehrhart-Bornstein, M.; et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: Correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2012, 13, 514–522. [Google Scholar] [CrossRef]
  175. Mei, Z.; Wang, F.; Bhosle, A.; Dong, D.; Mehta, R.; Ghazi, A.; Zhang, Y.; Liu, Y.; Rinott, E.; Ma, S.; et al. Strain-specific gut microbial signatures in type 2 diabetes identified in a cross-cohort analysis of 8,117 metagenomes. Nat. Med. 2024, 30, 2265–2276. [Google Scholar] [CrossRef] [PubMed]
  176. Sanna, S.; Van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vila, A.V.; Võsa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.M.A.E.; Oosting, M.; et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef]
  177. Khan, S.; Maremanda, K.P.; Jena, G. Butyrate, a short-chain fatty acid and histone deacetylases inhibitor: Nutritional, physiological, and pharmacological aspects in diabetes. In Handbook of Nutrition, Diet, and Epigenetics; Springer International Publishing: Berlin/Heidelberg, Germany, 2017; pp. 793–807. [Google Scholar]
  178. Stefan, N.; Fritsche, A.; Weikert, C.; Boeing, H.; Joost, H.-G.; Häring, H.-U.; Schulze, M.B. Plasma fetuin-a levels and the risk of type 2 diabetes. Diabetes 2008, 57, 2762–2767. [Google Scholar] [CrossRef]
  179. Ong, K.-L.; O’COnnell, R.; Januszewski, A.S.; Jenkins, A.J.; Xu, A.; Sullivan, D.R.; Barter, P.J.; Scott, R.S.; Taskinen, M.-R.; Waldman, B.; et al. Baseline circulating FGF21 concentrations and increase after fenofibrate treatment predict more rapid glycemic progression in type 2 diabetes: Results from the field study. Clin. Chem. 2017, 63, 1261–1270. [Google Scholar] [CrossRef] [PubMed]
  180. Woo, Y.C.; Lee, C.H.; Fong, C.H.; Xu, A.; Tso, A.W.; Cheung, B.M.; Lam, K.S. Serum fibroblast growth factor 21 is a superior biomarker to other adipokines in predicting incident diabetes. Clin. Endocrinol. 2017, 86, 37–43. [Google Scholar] [CrossRef] [PubMed]
  181. Ding, E.L.; Song, Y.; Manson, J.E.; Hunter, D.J.; Lee, C.C.; Rifai, N.; Buring, J.E.; Gaziano, J.M.; Liu, S. Sex hormone–binding globulin and risk of type 2 diabetes in women and men. N. Engl. J. Med. 2009, 361, 1152–1163. [Google Scholar] [CrossRef]
  182. Kwak, S.H.; Park, K.S. Recent progress in genetic and epigenetic research on type 2 diabetes. Exp. Mol. Med. 2016, 48, e220. [Google Scholar] [CrossRef]
  183. Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 2011, 11, 85–97. [Google Scholar] [CrossRef] [PubMed]
  184. Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Martín-Rodríguez, A.; Martínez-Guardado, I.; Navarro-Jiménez, E.; Laborde-Cárdenas, C.C.; Tornero-Aguilera, J.F. The role of adipokines in health and disease. Biomedicines 2023, 11, 1290. [Google Scholar] [CrossRef]
  185. Kirichenko, T.V.; Markina, Y.V.; Bogatyreva, A.I.; Tolstik, T.V.; Varaeva, Y.R.; Starodubova, A.V. The role of adipokines in inflammatory mechanisms of obesity. Int. J. Mol. Sci. 2022, 23, 14982. [Google Scholar] [CrossRef]
  186. Arita, Y.; Kihara, S.; Ouchi, N.; Takahashi, M.; Maeda, K.; Miyagawa, J.I.; Hotta, K.; Shimomura, I.; Nakamura, T.; Miyaoka, K.; et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem. Biophys. Res. Commun. 1999, 257, 79–83. [Google Scholar] [CrossRef]
  187. Yamauchi, T.; Kamon, J.; Minokoshi, Y.; Ito, Y.; Waki, H.; Uchida, S.; Yamashita, S.; Noda, M.; Kita, S.; Ueki, K.; et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med. 2002, 8, 1288–1295. [Google Scholar] [CrossRef]
  188. Feng, R.; Li, Y.; Wang, C.; Luo, C.; Liu, L.; Chuo, F.; Li, Q.; Sun, C. Higher vaspin levels in subjects with obesity and type 2 diabetes mellitus: A meta-analysis. Diabetes Res. Clin. Pract. 2014, 106, 88–94. [Google Scholar] [CrossRef]
  189. Youn, B.-S.; KlötIng, N.; Kratzsch, J.; Lee, N.; Park, J.W.; Song, E.-S.; Ruschke, K.; Oberbach, A.; Fasshauer, M.; Stumvoll, M.; et al. Serum vaspin concentrations in human obesity and type 2 diabetes. Diabetes 2008, 57, 372–377. [Google Scholar] [CrossRef] [PubMed]
  190. Lesná, J.; Tichá, A.; Hyšpler, R.; Musil, F.; Bláha, V.; Sobotka, L.; Zadák, Z.; Šmahelová, A. Omentin-1 plasma levels and cholesterol metabolism in obese patients with diabetes mellitus type 1: Impact of weight reduction. Nutr. Diabetes 2015, 5, e183. [Google Scholar] [CrossRef] [PubMed]
  191. Hedjazifar, S.; Shahidi, R.K.; Hammarstedt, A.; Bonnet, L.; Church, C.; Boucher, J.; Blüher, M.; Smith, U. The novel adipokine gremlin 1 antagonizes insulin action and is increased in type 2 diabetes and NAFLD/NASH. Diabetes 2020, 69, 331–341. [Google Scholar] [CrossRef]
  192. Post, A.; Dam, W.; Sokooti, S.; Groothof, D.; Gloerich, J.; van Gool, A.J.; Kremer, D.; Gansevoort, R.T.; Van den Born, J.; Kema, I.P.; et al. Circulating FGF21 concentration, fasting plasma glucose, and the risk of type 2 diabetes: Results from the PREVEND study. J. Clin. Endocrinol. Metab. 2023, 108, 1387–1393. [Google Scholar] [CrossRef] [PubMed]
  193. Kharitonenkov, A.; Shiyanova, T.L.; Koester, A.; Ford, A.M.; Micanovic, R.; Galbreath, E.J.; Sandusky, G.E.; Hammond, L.J.; Moyers, J.S.; Owens, R.A.; et al. FGF-21 as a novel metabolic regulator. J. Clin. Investig. 2005, 115, 1627–1635. [Google Scholar] [CrossRef]
  194. Matsubara, M.; Maruoka, S.; Katayose, S. Inverse relationship between plasma adiponectin and leptin concentrations in normal-weight and obese women. Eur. J. Endocrinol. 2002, 147, 173–180. [Google Scholar] [CrossRef]
  195. Steppan, C.M.; Bailey, S.T.; Bhat, S.; Brown, E.J.; Banerjee, R.R.; Wright, C.M.; Patel, H.R.; Ahima, R.S.; Lazar, M.A. The hormone resistin links obesity to diabetes. Nature 2001, 409, 307–312. [Google Scholar] [CrossRef]
  196. Chen, B.H.; Song, Y.; Ding, E.L.; Roberts, C.K.; Manson, J.E.; Rifai, N.; Buring, J.E.; Gaziano, J.M.; Liu, S. Circulating levels of resistin and risk of type 2 diabetes in men and women: Results from two prospective cohorts. Diabetes Care 2009, 32, 329–334. [Google Scholar] [CrossRef]
  197. Mukherji, A.B.; Idowu, V.; Zhao, L.; Leung, L.L.K.; Shen, S.; Palaniappan, L.; Morser, J. Chemerin Levels in Individuals with Type 2 Diabetes and a Normal Weight versus Individuals with Type 2 Diabetes and Obesity: An Observational, Cross-Sectional Study. Biomedicines 2024, 12, 983. [Google Scholar] [CrossRef]
  198. Goralski, K.B.; McCarthy, T.C.; Hanniman, E.A.; Zabel, B.A.; Butcher, E.C.; Parlee, S.D.; Muruganandan, S.; Sinal, C.J. Chemerin, a novel adipokine that regulates adipogenesis and adipocyte metabolism. J. Biol. Chem. 2007, 282, 28175–28188. [Google Scholar] [CrossRef] [PubMed]
  199. Chen, M.-P.; Chung, F.-M.; Chang, D.-M.; Tsai, J.C.-R.; Huang, H.-F.; Shin, S.-J.; Lee, Y.-J. Elevated plasma level of visfatin/pre-B cell colony-enhancing factor in patients with type 2 diabetes mellitus. J. Clin. Endocrinol. Metab. 2006, 91, 295–299. [Google Scholar] [CrossRef]
  200. Graham, T.E.; Yang, Q.; Blüher, M.; Hammarstedt, A.; Ciaraldi, T.P.; Henry, R.R.; Wason, C.J.; Oberbach, A.; Jansson, P.-A.; Smith, U.; et al. Retinol-binding protein 4 and insulin resistance in lean, obese, and diabetic subjects. N. Engl. J. Med. 2006, 354, 2552–2563. [Google Scholar] [CrossRef]
  201. Hotamisligil, G.S.; Murray, D.L.; Choy, L.N.; Spiegelman, B.M. Tumor necrosis factor alpha inhibits signaling from the insulin receptor. Proc. Natl. Acad. Sci. USA 1994, 91, 4854–4858. [Google Scholar] [CrossRef]
  202. Qu, D.; Liu, J.; Lau, C.W.; Huang, Y. IL-6 in diabetes and cardiovascular complications. Br. J. Pharmacol. 2014, 171, 3595–3603. [Google Scholar] [CrossRef]
  203. Zampetaki, A.; Kiechl, S.; Drozdov, I.; Willeit, P.; Mayr, U.; Prokopi, M.; Mayr, A.; Weger, S.; Oberhollenzer, F.; Bonora, E.; et al. Plasma MicroRNA profiling reveals loss of endothelial MiR-126 and other MicroRNAs in type 2 diabetes. Circ. Res. 2010, 107, 810–817. [Google Scholar] [CrossRef]
  204. Thomson, J.M.; Parker, J.; Perou, C.M.; Hammond, S.M. A custom microarray platform for analysis of microRNA gene expression. Nat. Methods 2004, 1, 47–53. [Google Scholar] [CrossRef] [PubMed]
  205. Witwer, K.W.; McAlexander, M.A.; Queen, S.E.; Adams, R.J. Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: Limited evidence for general uptake of dietary plant xenomiRs. RNA Biol. 2013, 10, 1080–1086. [Google Scholar] [CrossRef] [PubMed]
  206. Niu, Y.; Zhang, L.; Qiu, H.; Wu, Y.; Wang, Z.; Zai, Y.; Liu, L.; Qu, J.; Kang, K.; Gou, D. An improved method for detecting circulating microRNAs with S-Poly(T) Plus real-time PCR. Sci. Rep. 2015, 5, 15100. [Google Scholar] [CrossRef]
  207. Cheng, Y.; Zhang, X.; Li, Z.; Jiao, X.; Wang, Y.; Zhang, Y. Highly sensitive determination of microRNA using target-primed and branched rolling-circle amplification. Angew. Chem. Int. Ed. Engl. 2009, 48, 3268–3272. [Google Scholar] [CrossRef]
  208. Yao, B.; Li, J.; Huang, H.; Sun, C.; Wang, Z.; Fan, Y.; Chang, Q.; Li, S.; Xi, J. Quantitative analysis of zeptomole microRNAs based on isothermal ramification amplification. RNA 2009, 15, 1787–1794. [Google Scholar] [CrossRef] [PubMed][Green Version]
  209. Yan, J.; Li, Z.; Liu, C.; Cheng, Y. Simple and sensitive detection of microRNAs with ligase chain reaction. Chem. Commun. 2010, 46, 2432–2434. [Google Scholar] [CrossRef] [PubMed]
  210. Miller, M.B.; Tang, Y.-W. Basic concepts of microarrays and potential applications in clinical microbiology. Clin. Microbiol. Rev. 2009, 22, 611–633. [Google Scholar] [CrossRef]
  211. Cao, X.; Chen, C.; Zhu, Q. Biosensors based on functional nucleic acids and isothermal amplification techniques. Talanta 2023, 253, 123977. [Google Scholar] [CrossRef]
  212. Gibriel, A.A.; Adel, O. Advances in ligase chain reaction and ligation-based amplifications for genotyping assays: Detection and applications. Mutat. Res. Mol. Mech. Mutagen. 2017, 773, 66–90. [Google Scholar] [CrossRef]
  213. Eggins, B.R. Biosensors: An Introduction; Springer-Verlag: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  214. Chambers, J.P.; Arulanandam, B.P.; Matta, L.L.; Weis, A.; Valdes, J.J. Biosensor recognition elements. Curr. Issues Mol. Biol. 2008, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  215. Iqbal, S.S.; Mayo, M.W.; Bruno, J.G.; Bronk, B.V.; A Batt, C.; Chambers, J.P. A review of molecular recognition technologies for detection of biological threat agents. Biosens. Bioelectron. 2000, 15, 549–578. [Google Scholar] [CrossRef]
  216. Malhotra, B.D.; Ali, M.A. Nanomaterials in Biosensors: Fundamentals and Applications; Springer-Verlag: Berlin/Heidelberg, Germany, 2017; pp. 1–74. [Google Scholar]
  217. Teniou, A.; Rhouati, A.; Marty, J.-L. Recent Advances in Biosensors for Diagnosis of Autoimmune Diseases. Sensors 2024, 24, 1510. [Google Scholar] [CrossRef]
  218. Baranwal, A.; Polash, S.A.; Aralappanavar, V.K.; Behera, B.K.; Bansal, V.; Shukla, R. Recent Progress and Prospect of Metal–Organic Framework-Based Nanozymes in Biomedical Application. Nanomaterials 2024, 14, 244. [Google Scholar] [CrossRef]
  219. Baranwal, A.; Chandra, P. Clinical implications and electrochemical biosensing of monoamine neurotransmitters in body fluids, in vitro, in vivo, and ex vivo models. Biosens. Bioelectron. 2018, 121, 137–152. [Google Scholar] [CrossRef]
  220. Chalklen, T.; Jing, Q.; Kar-Narayan, S. Biosensors based on mechanical and electrical detection techniques. Sensors 2020, 20, 5605. [Google Scholar] [CrossRef]
  221. Clark, L.C., Jr.; Lyons, C. Electrode systems for continuous monitoring in cardiovascular surgery. Ann. N. Y. Acad. Sci. 1962, 102, 29–45. [Google Scholar] [CrossRef]
  222. Updike, S.J.; Hicks, G.P. The enzyme electrode. Nature 1967, 214, 986–988. [Google Scholar] [CrossRef]
  223. Updike, S.J.; Hicks, G.P. Reagentless substrate analysis with immobilized enzymes. Science 1967, 158, 270–272. [Google Scholar] [CrossRef] [PubMed]
  224. Guilbault, G.; Lubrano, G. An enzyme electrode for the amperometric determination of glucose. Anal. Chim. Acta 1973, 64, 439–455. [Google Scholar] [CrossRef] [PubMed]
  225. Wang, J. Electrochemical glucose biosensors. Chem. Rev. 2008, 108, 814–825. [Google Scholar] [CrossRef]
  226. Yoo, E.-H.; Lee, S.-Y. Glucose biosensors: An overview of use in clinical practice. Sensors 2010, 10, 4558–4576. [Google Scholar] [CrossRef]
  227. Cass, A.E.G.; Davis, G.; Francis, G.D.; Hill, H.A.O.; Aston, W.J.; Higgins, I.J.; Plotkin, E.V.; Scott, L.D.L.; Turner, A.P.F. Ferrocene-mediated enzyme electrode for amperometric determination of glucose. Anal. Chem. 1984, 56, 667–671. [Google Scholar] [CrossRef]
  228. Chaubey, A.; Malhotra, B. Mediated biosensors. Biosens. Bioelectron. 2002, 17, 441–456. [Google Scholar] [CrossRef] [PubMed]
  229. Shichiri, M.; Yamasaki, Y.; Kawamori, R.; Hakui, N.; Abe, H. Wearable artificial endocrine pancreas with needle-type glucose sensor. Lancet 1982, 320, 1129–1131. [Google Scholar] [CrossRef]
  230. Frew, J.E.; Hill, H.A.O. Electrochemical biosensors. Anal. Chem. 1987, 59, 933A–944A. [Google Scholar] [CrossRef]
  231. Matthews, D.; Bown, E.; Watson, A.; Holman, R.; Steemson, J.; Hughes, S.; Scott, D. Pen-sized digital 30-second blood glucose meter. Lancet 1987, 329, 778–779. [Google Scholar] [CrossRef]
  232. Khan, G.F.; Ohwa, M.; Wernet, W. Design of a stable charge transfer complex electrode for a third-generation amperometric glucose sensor. Anal. Chem. 1996, 68, 2939–2945. [Google Scholar] [CrossRef]
  233. Palmisano, F.; Zambonin, P.G.; Centonze, D.; Quinto, M. A Disposable, reagentless, third-generation glucose biosensor based on overoxidized poly(pyrrole)/tetrathiafulvalene−tetracyanoquinodimethane composite. Anal. Chem. 2002, 74, 5913–5918. [Google Scholar] [CrossRef] [PubMed]
  234. Mastrototaro, J.J. The MiniMed continuous glucose monitoring system. Diabetes Technol. Ther. 2000, 2 (Suppl. S1), 13–18. [Google Scholar] [CrossRef]
  235. Cox, M. An overview of continuous glucose monitoring systems. J. Pediatr. Health Care 2009, 23, 344–347. [Google Scholar] [CrossRef] [PubMed]
  236. Csoeregi, E.; Schmidtke, D.W.; Heller, A. design and optimization of a selective subcutaneously implantable glucose electrode based on “wired” glucose oxidase. Anal. Chem. 1995, 67, 1240–1244. [Google Scholar] [CrossRef] [PubMed]
  237. Schmidtke, D.W.; Freeland, A.C.; Heller, A.; Bonnecaze, R.T. Measurement and modeling of the transient difference between blood and subcutaneous glucose concentrations in the rat after injection of insulin. Proc. Natl. Acad. Sci. USA 1998, 95, 294–299. [Google Scholar] [CrossRef]
  238. Klonoff, D.C. Noninvasive blood glucose monitoring. Diabetes Care 1997, 20, 433–437. [Google Scholar] [CrossRef]
  239. Goetz, M.; Cote, G.; Erckens, R.; March, W.; Motamedi, M. Application of a multivariate technique to Raman spectra for quantification of body chemicals. IEEE Trans. Biomed. Eng. 1995, 42, 728–731. [Google Scholar] [CrossRef] [PubMed]
  240. Rabinovitch, B.; March, W.F.; Adams, R.L. Noninvasive glucose monitoring of the aqueous humor of the eye: Part I. measurement of very small optical rotations. Diabetes Care 1982, 5, 254–258. [Google Scholar] [CrossRef] [PubMed]
  241. MacKenzie, H.; Ashton, H.S.; Spiers, S.; Shen, Y.; Freeborn, S.S.; Hannigan, J.; Lindberg, J.; Rae, P. Advances in photoacoustic noninvasive glucose testing. Clin. Chem. 1999, 45, 1587–1595. [Google Scholar] [CrossRef]
  242. Gabriely, I.; Wozniak, R.; Mevorach, M.; Kaplan, J.; Aharon, Y.; Shamoon, H. Transcutaneous glucose measurement using near-infrared spectroscopy during hypoglycemia. Diabetes Care 1999, 22, 2026–2032. [Google Scholar] [CrossRef] [PubMed]
  243. Larin, K.V.; Eledrisi, M.S.; Motamedi, M.; Esenaliev, R.O. Noninvasive blood glucose monitoring with optical coherence tomography: A pilot study in human subjects. Diabetes Care 2002, 25, 2263–2267. [Google Scholar] [CrossRef]
  244. Tierney, M.; Tamada, J.; Potts, R.; Jovanovic, L.; Garg, S. Clinical evaluation of the GlucoWatch® biographer: A continual, non-invasive glucose monitor for patients with diabetes. Biosens. Bioelectron. 2001, 16, 621–629. [Google Scholar] [CrossRef]
  245. Corabian, P.; Chojecki, D. Exploratory Brief on Glucose Monitoring Technologies; Institute of Health Economics: Edmonton, AB, Canada, 2019. [Google Scholar]
  246. Kropff, J.; Choudhary, P.; Neupane, S.; Barnard, K.; Bain, S.C.; Kapitza, C.; Forst, T.; Link, M.; Dehennis, A.; DeVries, J.H. Accuracy and longevity of an implantable continuous glucose sensor in the PRECISE study: A 180-day, prospective, multicenter, pivotal trial. Diabetes Care 2017, 40, 63–68. [Google Scholar] [CrossRef]
  247. Boscari, F.; Vettoretti, M.; Cavallin, F.; Amato, A.M.L.; Uliana, A.; Vallone, V.; Avogaro, A.; Facchinetti, A.; Bruttomesso, D. Implantable and transcutaneous continuous glucose monitoring system: A randomized cross over trial comparing accuracy, efficacy and acceptance. J. Endocrinol. Investig. 2021, 45, 115–124. [Google Scholar] [CrossRef]
  248. Harb, F.; Azar, W.S.; Ghadieh, H.E.; Njeim, R.; Tawk, Y.; Costantine, J.; Kanj, R.; Eid, A.A. Future Developments in Invasive and Non-invasive Diabetes Monitoring. In Advanced Bioscience and Biosystems for Detection and Management of Diabetes; Springer: Berlin/Heidelberg, Germany, 2022; pp. 293–313. [Google Scholar]
  249. Irfani, M.Z.; Koesoema, A.P. Continuous and Non-Invasive Blood Glucose Measurements: A Narrative Review. In Proceedings of the 2023 10th International Conference on Information Technology, Computer, and Electrical Engineering (ICITACEE), Semarang, Indonesia, 31 August 2023–1 September 2023. [Google Scholar]
  250. Lee, I.; Probst, D.; Klonoff, D.; Sode, K. Continuous glucose monitoring systems—Current status and future perspectives of the flagship technologies in biosensor research. Biosens. Bioelectron. 2021, 181, 113054. [Google Scholar] [CrossRef]
  251. Research, G.V. Glucose Biosensors Market Size & Trends. In Glucose Biosensors Market Size, Share & Trends Analysis Report By Type (Electrochemical Biosensors, Optical Biosensors), by End Use (Hospitals, Homecare, Diagnostic Centers), by Region, and Segment Forecasts, 2025–2030; Grand View Research, Inc.: San Francisco, CA, USA, 2024; p. 190. [Google Scholar]
  252. Patil, R. Glucose Biosensors Market Size, Share, and Trends 2025 to 2034; Shivarkar, A., Ed.; Precedence Research: Ottawa, ON, Canada, 2024. [Google Scholar]
  253. Benjamin, E.M. Self-monitoring of blood glucose: The basics. Clin. Diabetes 2002, 20, 45–47. [Google Scholar] [CrossRef]
  254. Huang, X.; Leduc, C.; Ravussin, Y.; Li, S.; Davis, E.; Song, B.; Wang, Q.; Accili, D.; Leibel, R.; Lin, Q. Continuous monitoring of glucose in subcutaneous tissue using microfabricated differential affinity sensors. J. Diabetes Sci. Technol. 2012, 6, 1436–1444. [Google Scholar] [CrossRef]
  255. Malik, S.; Singh, J.; Goyat, R.; Saharan, Y.; Chaudhry, V.; Umar, A.; Ibrahim, A.A.; Akbar, S.; Ameen, S.; Baskoutas, S. Nanomaterials-based biosensor and their applications: A review. Heliyon 2023, 9, e19929. [Google Scholar] [CrossRef]
  256. Sannino, D. Types and classification of nanomaterials. In Nanotechnology: Trends and Future Applications; Springer: Singapore, 2021; pp. 15–38. [Google Scholar]
  257. Graybill, R.M.; Bailey, R.C. Emerging biosensing approaches for microRNA analysis. Anal. Chem. 2016, 88, 431–450. [Google Scholar] [CrossRef]
  258. Johnson, B.N.; Mutharasan, R. Biosensor-based microRNA detection: Techniques, design, performance, and challenges. Analyst 2014, 139, 1576–1588. [Google Scholar] [CrossRef] [PubMed]
  259. Khandan-Nasab, N.; Askarian, S.; Mohammadinejad, A.; Aghaee-Bakhtiari, S.H.; Mohajeri, T.; Oskuee, R.K. Biosensors, microfluidics systems and lateral flow assays for circulating microRNA detection: A review. Anal. Biochem. 2021, 633, 114406. [Google Scholar] [CrossRef]
  260. Lin, X.; Wang, K.; Luo, C.; Yang, M.; Wu, J. MicroRNA biosensors for early detection of hepatocellular carcinoma. Chemosensors 2023, 11, 504. [Google Scholar] [CrossRef]
  261. Kilic, T.; Erdem, A.; Ozsoz, M.; Carrara, S. microRNA biosensors: Opportunities and challenges among conventional and commercially available techniques. Biosens. Bioelectron. 2018, 99, 525–546. [Google Scholar] [CrossRef]
  262. Liu, W.-J.; Wang, L.-J.; Zhang, C.-Y. Progress in quantum dot-based biosensors for microRNA assay: A review. Anal. Chim. Acta 2023, 1278, 341615. [Google Scholar] [CrossRef]
  263. Hanoglu, S.B.; Harmanci, D.; Ucar, N.; Evran, S.; Timur, S. Recent approaches in magnetic nanoparticle-based biosensors of miRNA detection. Magnetochemistry 2023, 9, 23. [Google Scholar] [CrossRef]
  264. Gorgani, L.; Mohammadi, M.; Darzi, G.N.; Raoof, J.B. Metal-organic framework (MOF)-based biosensors for miRNA detection. Talanta 2024, 273, 125854. [Google Scholar] [CrossRef]
  265. Esmaeilzadeh, A.A.; Yaseen, M.M.; Khudaynazarov, U.; Al-Gazally, M.E.; Opulencia, M.J.C.; Jalil, A.T.; Mohammed, R.N. Recent advances on the electrochemical and optical biosensing strategies for monitoring microRNA-21: A review. Anal. Methods 2022, 14, 4449–4459. [Google Scholar] [CrossRef]
  266. Lu, X.; Yao, C.; Sun, L.; Li, Z. Plasmon-enhanced biosensors for microRNA analysis and cancer diagnosis. Biosens. Bioelectron. 2022, 203, 114041. [Google Scholar] [CrossRef]
  267. Meng, X.; Pang, X.; Yang, J.; Zhang, X.; Dong, H. Recent advances in electrochemiluminescence biosensors for MicroRNA detection. Small 2024, 20, e2307701. [Google Scholar] [CrossRef]
  268. Tran, H.V.; Piro, B. Recent trends in application of nanomaterials for the development of electrochemical microRNA biosensors. Microchim. Acta 2021, 188, 128. [Google Scholar] [CrossRef]
  269. Negahdary, M.; Angnes, L. Application of electrochemical biosensors for the detection of microRNAs (miRNAs) related to cancer. Chem. Rev. 2022, 464, 214565. [Google Scholar] [CrossRef]
  270. Wang, Z.-Y.; Sun, M.-H.; Zhang, Q.; Li, P.-F.; Wang, K.; Li, X.-M. Advances in Point-of-Care Testing of microRNAs Based on Portable Instruments and Visual Detection. Biosensors 2023, 13, 747. [Google Scholar] [CrossRef]
  271. Song, S.; Guo, Y.; Mao, D.; Gao, H.; Gao, Y.-P.; Kang, W. An ultrasensitive electrochemical/colorimetric dual-mode self-powered biosensing platform for lung cancer marker detection by multiple-signal amplification strategy. Anal. Chim. Acta 2024, 1316, 342827. [Google Scholar] [CrossRef] [PubMed]
  272. Hou, Y.-Y.; Xie, W.-Z.; Tan, X.; Huang, K.-J.; Xu, J. Superior graphdiyne self-powered biosensing platform with highly sensitivity and reliability for dual-mode detection of MicroRNA by integrating T7 Exonuclease and 3D DNA walker induced rolling circle amplification. Anal. Chim. Acta 2023, 1239, 340696. [Google Scholar] [CrossRef]
  273. Ma, Y.; Shi, J.; Lin, Y.; Wu, Y.; Luo, H.; Yan, J.; Huang, K.-J.; Tan, X. Smart enzyme-free amplification dual-mode self-powered platform designed on two-dimensional networked graphdiyne and DNA nanorods for ultra-sensitive detection of breast cancer biomarkers. Anal. Chim. Acta 2023, 1280, 341876. [Google Scholar] [CrossRef] [PubMed]
  274. Wang, Y.-Z.; Wang, L.-L.; Liu, Y.; Zhang, Y.-Q.; Li, M.-L.; Chen, C.-X.; Zhu, J.-W.; Yang, F.; Hu, Y.-H. Dual “on-off” signal conversion strategy based on surface plasmon coupling and resonance energy transfer for visual electrochemiluminescence ratiometric analysis of MiRNA-141. Biosens. Bioelectron. 2024, 253, 116162. [Google Scholar] [CrossRef] [PubMed]
  275. Zoughi, S.; Faridbod, F.; Moradi, S. Rapid enzyme-free detection of miRNA-21 in human ovarian cancerous cells using a fluorescent nanobiosensor designed based on hairpin DNA-templated silver nanoclusters. Anal. Chim. Acta 2024, 1320, 342968. [Google Scholar] [CrossRef]
  276. Dargah, M.M.; Youseftabar-Miri, L.; Divsar, F.; Hosseinjani-Pirdehi, H.; Mahani, M.; Bakhtiari, S.; Montazar, L. Triplex hairpin oligosensor for ultrasensitive determination of miRNA-155 as a cancer marker using Si quantum dots and Au nanoparticles. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2024, 322, 124750. [Google Scholar] [CrossRef] [PubMed]
  277. Liu, X.; Wang, Q.; Diao, Z.; Huo, D.; Hou, C. Label-free fluorescent biosensor based on AuNPs etching releasing signal for miRNA-155 detection. Talanta 2024, 278, 126481. [Google Scholar] [CrossRef]
  278. Afzalinia, A.; Mirzaee, M. Ultrasensitive fluorescent miRNA biosensor based on a “sandwich” oligonucleotide hybridization and fluorescence resonance energy transfer process using an Ln(III)-MOF and Ag nanoparticles for early cancer diagnosis: Application of central composite design. ACS Appl. Mater. Interfaces 2020, 12, 16076–16087. [Google Scholar] [CrossRef]
  279. Qin, Z.; Fu, J.; Wang, J.; Deng, S.; Xiong, F.; Gao, Q.; Ye, J.; Zhang, Y.; Li, S. An intelligent fluorescence sensing platform based on entropy-driven toehold-mediated strand displacement cycle reaction for point-of-care testing of miRNA. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2025, 325, 125177. [Google Scholar] [CrossRef]
  280. Li, Y.; Tu, C.; Chen, Q.; Lin, Y.; Li, B.; Lyu, H. Enhanced red emission of upconversion nanoparticles via Li+ and Tm3+ codoping and active core-shell construction for sensitive detection of miRNAs. Anal. Chim. Acta 2025, 1335, 343429. [Google Scholar] [CrossRef]
  281. Jia, Z.; Tu, K.; Xu, Q.; Gao, W.; Liu, C.; Fang, B.; Zhang, M. A novel disease-associated nucleic acid sensing platform based on split DNA-scaffolded sliver nanocluster. Anal. Chim. Acta 2021, 1175, 338734. [Google Scholar] [CrossRef]
  282. Li, P.; Ye, Y.; Li, Y.; Xie, Z.; Ye, L.; Huang, J. A MoS2 nanosheet-based CRISPR/Cas12a biosensor for efficient miRNA quantification for acute myocardial infarction. Biosens. Bioelectron. 2024, 251, 116129. [Google Scholar] [CrossRef] [PubMed]
  283. Ma, F.; Jiang, S.; Zhang, C.-Y. SiRNA-directed self-assembled quantum dot biosensor for simultaneous detection of multiple microRNAs at the single-particle level. Biosens. Bioelectron. 2020, 157, 112177. [Google Scholar] [CrossRef]
  284. Yu, J.; Liu, Q.; Qi, L.; Fang, Q.; Shang, X.; Zhang, X.; Du, Y. Fluorophore and nanozyme-functionalized DNA walking: A dual-mode DNA logic biocomputing platform for microRNA sensing in clinical samples. Biosens. Bioelectron. 2024, 252, 116137. [Google Scholar] [CrossRef] [PubMed]
  285. Zhao, S.; Jia, Y.; Wang, A.; Yang, J.; Yang, L. Dual-mode DNA walker-based optical fiber biosensor for ultrasensitive detection of microRNAs. Biosens. Bioelectron. 2023, 239, 115613. [Google Scholar] [CrossRef] [PubMed]
  286. Fakhri, N.; Abarghoei, S.; Dadmehr, M.; Hosseini, M.; Sabahi, H.; Ganjali, M.R. Paper based colorimetric detection of miRNA-21 using Ag/Pt nanoclusters. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 227, 117529. [Google Scholar] [CrossRef]
  287. Zeng, Y.; Yue, H.; Cao, B.; Li, Y.; Yang, M.; Mao, C. Target-triggered formation of artificial enzymes on filamentous phage for ultrasensitive direct detection of circulating miRNA biomarkers in clinical samples. Angew. Chem. Int. Ed. Engl. 2022, 61, e202210121. [Google Scholar] [CrossRef]
  288. Aamri, M.; Mohammadi, H.; Amine, A. A highly sensitive colorimetric DNA sensor for MicroRNA-155 detection: Leveraging the peroxidase-like activity of copper nanoparticles in a double amplification strategy. Microchim. Acta 2024, 191, 32. [Google Scholar] [CrossRef]
  289. Shahsavar, K.; Shokri, E.; Hosseini, M. Sensitive colorimetric detection of miRNA-155 via G-quadruplex DNAzyme decorated spherical nucleic acid. Microchim. Acta 2022, 189, 357. [Google Scholar] [CrossRef]
  290. Cai, J.; Ding, L.; Gong, P.; Huang, J. A colorimetric detection of microRNA-148a in gastric cancer by gold nanoparticle–RNA conjugates. Nanotechnology 2019, 31, 095501. [Google Scholar] [CrossRef] [PubMed]
  291. An, J.; Zhang, M.; Fu, Y.; Zhang, Q.; Si, Y.; Zhang, Y.; Fang, Y.; Zhang, D. Emerging electrochemical biosensors for lung cancer-associated protein biomarker and miRNA detection. Int. J. Biol. Macromol. 2024, 280, 135972. [Google Scholar] [CrossRef]
  292. Pothipor, C.; Aroonyadet, N.; Bamrungsap, S.; Jakmunee, J.; Ounnunkad, K. A highly sensitive electrochemical microRNA-21 biosensor based on intercalating methylene blue signal amplification and a highly dispersed gold nanoparticles/graphene/polypyrrole composite. Analyst 2021, 146, 2679–2688. [Google Scholar] [CrossRef]
  293. Ebrahimi, A.; Nikokar, I.; Zokaei, M.; Bozorgzadeh, E. Design, development and evaluation of microRNA-199a-5p detecting electrochemical nanobiosensor with diagnostic application in Triple Negative Breast Cancer. Talanta 2018, 189, 592–598. [Google Scholar] [CrossRef] [PubMed]
  294. Sargazi, S.; Fatima, I.; Kiani, M.H.; Mohammadzadeh, V.; Arshad, R.; Bilal, M.; Rahdar, A.; Díez-Pascual, A.M.; Behzadmehr, R. Fluorescent-based nanosensors for selective detection of a wide range of biological macromolecules: A comprehensive review. Int. J. Biol. Macromol. 2022, 206, 115–147. [Google Scholar] [CrossRef]
  295. Dong, H.; Zhang, J.; Ju, H.; Lu, H.; Wang, S.; Jin, S.; Hao, K.; Du, H.; Zhang, X. Highly sensitive multiple microRNA detection based on fluorescence quenching of graphene oxide and isothermal strand-displacement polymerase reaction. Anal. Chem. 2012, 84, 4587–4593. [Google Scholar] [CrossRef]
  296. Mohammadi, S.; Salimi, A. Fluorometric determination of microRNA-155 in cancer cells based on carbon dots and MnO2 nanosheets as a donor-acceptor pair. Microchim. Acta 2018, 185, 372. [Google Scholar] [CrossRef] [PubMed]
  297. Peng, J.; Li, B.; Ma, Z.; Qiu, Z.; Hu, H.; Jiang, Y.; Gao, D. A microfluidic-based chemiluminescence biosensor for sensitive multiplex detection of exosomal microRNAs based on hybridization chain reaction. Talanta 2025, 281, 126838. [Google Scholar] [CrossRef] [PubMed]
  298. Bi, S.; Chen, M.; Jia, X.; Dong, Y. A hot-spot-active magnetic graphene oxide substrate for microRNA detection based on cascaded chemiluminescence resonance energy transfer. Nanoscale 2015, 7, 3745–3753. [Google Scholar] [CrossRef] [PubMed]
  299. Zhang, H.-R.; Xu, J.-J.; Chen, H.-Y. Electrochemiluminescence ratiometry: A new approach to DNA biosensing. Anal. Chem. 2013, 85, 5321–5325. [Google Scholar] [CrossRef]
  300. Coutinho, C.; Somoza, Á. MicroRNA sensors based on gold nanoparticles. Anal. Bioanal. Chem. 2019, 411, 1807–1824. [Google Scholar] [CrossRef]
  301. Gao, X.; Xu, L.-P.; Wu, T.; Wen, Y.; Ma, X.; Zhang, X. An enzyme-amplified lateral flow strip biosensor for visual detection of microRNA-224. Talanta 2016, 146, 648–654. [Google Scholar] [CrossRef]
  302. Peng, W.; Zhao, Q.; Piao, J.; Zhao, M.; Huang, Y.; Zhang, B.; Gao, W.; Zhou, D.; Shu, G.; Gong, X.; et al. Ultra-sensitive detection of microRNA-21 based on duplex-specific nuclease-assisted target recycling and horseradish peroxidase cascading signal amplification. Sens. Actuators B Chem. 2018, 263, 289–297. [Google Scholar] [CrossRef]
  303. Pitou, M.; Papi, R.M.; Tzavellas, A.-N.; Choli-Papadopoulou, T. ssDNA-Modified Gold Nanoparticles as a Tool to Detect miRNA Biomarkers in Osteoarthritis. ACS Omega 2023, 8, 7529–7535. [Google Scholar] [CrossRef]
  304. Shi, H.-Y.; Yang, L.; Zhou, X.-Y.; Bai, J.; Gao, J.; Jia, H.-X.; Li, Q.-G. A gold nanoparticle-based colorimetric strategy coupled to duplex-specific nuclease signal amplification for the determination of microRNA. Microchim. Acta 2017, 184, 525–531. [Google Scholar] [CrossRef]
  305. Baranwal, A.; Shukla, R.; Bansal, V. Nanozyme-enhanced paper-based biosensor technologies. TrAC Trends Anal. Chem. 2024, 172, 117573. [Google Scholar] [CrossRef]
  306. Wu, J.; Wang, X.; Wang, Q.; Lou, Z.; Li, S.; Zhu, Y.; Qin, L.; Wei, H. Nanomaterials with enzyme-like characteristics (nanozymes): Next-generation artificial enzymes (II). Chem. Soc. Rev. 2019, 48, 1004–1076. [Google Scholar] [CrossRef]
  307. Zhao, H.; Qu, Y.; Yuan, F.; Quan, X. A visible and label-free colorimetric sensor for miRNA-21 detection based on peroxidase-like activity of graphene/gold-nanoparticle hybrids. Anal. Methods 2016, 8, 2005–2012. [Google Scholar] [CrossRef]
  308. Yan, C.; Guo, J.; Qu, X.; Sun, H.; Chai, H.; Miao, P. Distance-Based Visual miRNA Biosensor with Strand Displacement Amplification-Mediated DNA Hydrogel Assembly. ACS Mater. Lett. 2024, 6, 2111–2117. [Google Scholar] [CrossRef]
  309. Li, D.; Cheng, W.; Yan, Y.; Zhang, Y.; Yin, Y.; Ju, H.; Ding, S. A colorimetric biosensor for detection of attomolar microRNA with a functional nucleic acid-based amplification machine. Talanta 2016, 146, 470–476. [Google Scholar] [CrossRef]
  310. Wang, C.; Zhang, Y.; Liu, C.; Gou, S.; Hu, S.; Guo, W. A portable colorimetric point-of-care testing platform for MicroRNA detection based on programmable entropy-driven dynamic DNA network modulated DNA-gold nanoparticle hybrid hydrogel film. Biosens. Bioelectron. 2023, 225, 115073. [Google Scholar] [CrossRef]
  311. Piao, J.; Zhao, Q.; Zhou, D.; Peng, W.; Gao, W.; Chen, M.; Shu, G.; Gong, X.; Chang, J. Enzyme-free colorimetric detection of MicroRNA-21 using metal chelator as label for signal generation and amplification. Anal. Chim. Acta 2019, 1052, 145–152. [Google Scholar] [CrossRef]
  312. Moabelo, K.L.; Lerga, T.M.; Jauset-Rubio, M.; Sibuyi, N.R.S.; O’sullivan, C.K.; Meyer, M.; Madiehe, A.M. A label-free gold nanoparticles-based optical aptasensor for the detection of retinol binding protein 4. Biosensors 2022, 12, 1061. [Google Scholar] [CrossRef] [PubMed]
  313. Torabi, R.; Ghourchian, H. Ultrasensitive nano-aptasensor for monitoring retinol binding protein 4 as a biomarker for diabetes prognosis at early stages. Sci. Rep. 2020, 10, 594. [Google Scholar] [CrossRef]
  314. Giorgi-Coll, S.; Marín, M.J.; Sule, O.; Hutchinson, P.J.; Carpenter, K.L. Aptamer-modified gold nanoparticles for rapid aggregation-based detection of inflammation: An optical assay for interleukin-6. Microchim. Acta 2020, 187, 13. [Google Scholar] [CrossRef]
  315. Kim, W.; Bang, A.; Kim, S.; Lee, G.-J.; Kim, Y.-H.; Choi, S. Adiponectin-targeted SERS immunoassay biosensing platform for early detection of gestational diabetes mellitus. Biosens. Bioelectron. 2022, 213, 114488. [Google Scholar] [CrossRef]
  316. He, J.; Wu, S.; Chen, W.; Kim, A.; Yang, W.; Wang, C.; Gu, Z.; Shen, J.; Dai, S.; Chen, W.; et al. Calligraphy of Nanoplasmonic Bioink-Based Multiplex Immunosensor for Precision Immune Monitoring and Modulation. ACS Appl. Mater. Interfaces 2023, 15, 50047–50057. [Google Scholar] [CrossRef]
  317. Li, H.; Li, X.; Meng, Q.; Han, J.; Zhao, W.; Chen, J.; Su, W.; Song, M.; Shi, C.; Wang, L. Electric field-induced alignment of Ag/Au nanowires for ultrasensitive in situ detection of Interleukin-6. Biosens. Bioelectron. 2025, 271, 117033. [Google Scholar] [CrossRef]
  318. Sytu, M.R.C.; Stoner, A.; Hahm, J.-I. Strain-Modulated and Nanorod-Waveguided Fluorescence in Single Zinc Oxide Nanorod-Based Immunodetection. Biosensors 2024, 14, 85. [Google Scholar] [CrossRef]
  319. Erkmen, C.; Tığ, G.A.; Uslu, B. Nanomaterial-based sandwich-type electrochemical aptasensor platform for sensitive voltammetric determination of leptin. Microchim. Acta 2022, 189, 396. [Google Scholar] [CrossRef]
  320. Zhang, Q.; Li, Y.; Hu, Q.; Xie, R.; Zhou, W.; Liu, X.; Wang, Y. Smartphone surface plasmon resonance imaging for the simultaneous and sensitive detection of acute kidney injury biomarkers with noninvasive urinalysis. Lab A Chip 2022, 22, 4941–4949. [Google Scholar] [CrossRef] [PubMed]
  321. Erkmen, C.; Tiğ, G.A.; Uslu, B. First label-free impedimetric aptasensor based on Au NPs/TiO2 NPs for the determination of leptin. Sens. Actuators B Chem. 2022, 358, 131420. [Google Scholar] [CrossRef]
  322. Islam, T.; Ahsan, A.; Hassan, M.; Afrin, H.; Pena-Zacarias, J.; Aldalbahi, A.; Alvarado-Tenorio, B.; Noveron, J.C. Nurunnabi detection of leptin using electrocatalyst mediated impedimetric sensing. ACS Biomater. Sci. Eng. 2022, 9, 2170–2180. [Google Scholar] [CrossRef] [PubMed]
  323. Qiu, Y.; Jiang, K.; Wu, J.; Mi, H.; Peng, Y.-K.; Go, Y.Y.; Park, H.J.; Lee, J.-H. Fast and sensitive immuno-PCR assisted by plasmonic magnetic nanoparticles. Appl. Mater. Today 2021, 23, 101054. [Google Scholar] [CrossRef]
  324. Li, A.; Mo, X.; Lu, Y.; Zhu, G.; Liu, C.; Yang, X.; Huang, Y.; Sheng, J.; Zhang, H.; Meng, D.; et al. Digital SERS immunoassay of Interleukin-6 based on Au@Ag-Au nanotags. Biosens. Bioelectron. 2025, 270, 116973. [Google Scholar] [CrossRef]
  325. Zhang, C.; Liu, L.; Li, H.; Hu, J.; Zhang, J.; Zhou, H.; Zhang, Z.; Du, X. An oriented antibody immobilization based electrochemical platform for detection of leptin in human with different body mass index. Sens. Actuators B Chem. 2022, 353, 131074. [Google Scholar] [CrossRef]
  326. Cai, J.; Gou, X.; Sun, B.; Li, W.; Li, D.; Liu, J.; Hu, F.; Li, Y. Porous graphene-black phosphorus nanocomposite modified electrode for detection of leptin. Biosens. Bioelectron. 2019, 137, 88–95. [Google Scholar] [CrossRef]
  327. Liu, Y.; Zhang, L.; Li, Q.; Dai, H.; Xiang, T.; Yang, G.; Li, L. A reusable colorimetric assay based on mixed valence state Ce-MOF@Pt nanoparticles for highly sensitive detection of visfatin. Anal. Chim. Acta 2021, 1146, 24–32. [Google Scholar] [CrossRef]
  328. Ali, M.; Sajid, M.; Khalid, M.A.U.; Kim, S.W.; Lim, J.H.; Huh, D.; Choi, K.H. A fluorescent lateral flow biosensor for the quantitative detection of Vaspin using upconverting nanoparticles. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 226, 117610. [Google Scholar] [CrossRef]
  329. Hosseini, Z.; Shi, D.; Yuan, J. A flexible multiplexed electrochemical biosensing platform with graphene and gold nanoparticle modification for enhanced e-ELISA point-of-care biomarker detection. Microchem. J. 2025, 208, 112437. [Google Scholar] [CrossRef]
  330. Liu, X.; Tseng, C.-L.; Lin, L.-Y.; Lee, C.-A.; Li, J.; Feng, L.; Song, L.; Li, X.; He, J.-H.; Sakthivel, R.; et al. Template-free synthesis of mesoporous Ce3NbO7/CeO2 hollow nanospheres for label-free electrochemical immunosensing of leptin. Sens. Actuators B Chem. 2021, 341, 130005. [Google Scholar] [CrossRef]
  331. Tertis, M.; Leva, P.I.; Bogdan, D.; Suciu, M.; Graur, F.; Cristea, C. Impedimetric aptasensor for the label-free and selective detection of Interleukin-6 for colorectal cancer screening. Biosens. Bioelectron. 2019, 137, 123–132. [Google Scholar] [CrossRef] [PubMed]
  332. Mahani, M.; Faghihi-Fard, M.; Divsar, F.; Torkzadeh-Mahani, M.; Khakbaz, F. Ultrasensitive FRET-based aptasensor for interleukin-6 as a biomarker for COVID-19 progression using nitrogen-doped carbon quantum dots and gold nanoparticles. Microchim. Acta 2022, 189, 472. [Google Scholar] [CrossRef]
  333. Gao, Z.; Wang, C.; He, J.; Chen, P. Pd@ Pt Nanodendrites as peroxidase nanomimics for enhanced colorimetric ELISA of cytokines with femtomolar sensitivity. Chemosensors 2022, 10, 359. [Google Scholar] [CrossRef] [PubMed]
  334. Adrover-Jaume, C.; Alba-Patiño, A.; Clemente, A.; Santopolo, G.; Vaquer, A.; Russell, S.M.; Barón, E.; Del Mar Gonzalez, M.; Ferrer, J.M.; Berman-Riu, M.; et al. Paper biosensors for detecting elevated IL-6 levels in blood and respiratory samples from COVID-19 patients. Sens. Actuators B Chem. 2021, 330, 129333. [Google Scholar] [CrossRef]
  335. Liang, B.; Wang, S.; Zheng, J.; Li, B.; Cheng, N.; Gan, N. All-in-one microfluidic immunosensing device for rapid and end-to-end determination of salivary biomarkers of cardiovascular diseases. Biosens. Bioelectron. 2025, 271, 117077. [Google Scholar] [CrossRef] [PubMed]
  336. Helmerhorst, E.; Chandler, D.J.; Nussio, M.; Mamotte, C.D. Real-time and label-free bio-sensing of molecular interactions by surface plasmon resonance: A laboratory medicine perspective. Clin. Biochem. Rev. 2012, 33, 161–173. [Google Scholar] [PubMed]
  337. Kukkar, D.; Chhillar, M.; Kim, K.-H. Application of SERS-based nanobiosensors to metabolite biomarkers of CKD. Biosens. Bioelectron. 2023, 232, 115311. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of types of diabetes. Among the three types, type 1, type 2 and gestational diabetes, type 2 is most common and contributes to an estimate of 90–95% of all diabetes cases.
Figure 1. Schematic illustration of types of diabetes. Among the three types, type 1, type 2 and gestational diabetes, type 2 is most common and contributes to an estimate of 90–95% of all diabetes cases.
Biosensors 15 00639 g001
Figure 2. Global diabetes demographics in 2021. Data is sourced from the IDF Diabetes Atlas 2021 [12].
Figure 2. Global diabetes demographics in 2021. Data is sourced from the IDF Diabetes Atlas 2021 [12].
Biosensors 15 00639 g002
Figure 3. Microvascular and macrovascular complications associated with diabetes.
Figure 3. Microvascular and macrovascular complications associated with diabetes.
Biosensors 15 00639 g003
Figure 4. Schematic illustration of a typical biosensor component.
Figure 4. Schematic illustration of a typical biosensor component.
Biosensors 15 00639 g004
Figure 5. Timeline of major milestones in glucose biosensor development.
Figure 5. Timeline of major milestones in glucose biosensor development.
Biosensors 15 00639 g005
Figure 6. The pie chart shows number of studies published on miRNA detection by employing biosensor (a) and nanobiosensor (b) platforms in the last ten years (2015–2024). The data was obtained from SCOPUS database by searching keywords microRNA* OR miRNA* AND biosens* for figure (a) and microRNA* OR miRNA* AND biosens* AND nano* for figure (b).
Figure 6. The pie chart shows number of studies published on miRNA detection by employing biosensor (a) and nanobiosensor (b) platforms in the last ten years (2015–2024). The data was obtained from SCOPUS database by searching keywords microRNA* OR miRNA* AND biosens* for figure (a) and microRNA* OR miRNA* AND biosens* AND nano* for figure (b).
Biosensors 15 00639 g006
Figure 7. Schematic illustration of EC nanobiosensor fabrication for miRNA sensing using single- (a) and dual-mode (EC/colourimetric) (b) detection. In single-mode sensor (a), a screen-printed carbon electrode (SPCE) modified with polypyrrole (PPY)-graphene nanocomposite, AuNP-conjugated ssDNA probe, and 6-mercapto-1-hexanol (MCH) acts as a sensor probe. When the target miRNA is present, methylene blue (MB) intercalates with the hybridised ssDNA probe-miRNA duplex, generating a strong peak current. In contrast, a significantly smaller peak current is observed in the absence of the target miRNA due to lesser intercalation of MB. The dual-mode sensor (b) employs carbon paper (CP) patterned with AuNP-vanadium sulphide nanosheet self-assembled nanorod (AuNP-VS4 NS NR) nanocomposite, modified with the H1 probe to function as the EC sensor probe. In parallel, the colourimetric sensor probe is created by modifying the AuNP-VS4 NS NR patterned CP with glucose oxidase (GOD). In the presence of target miRNA, a y-shaped DNA generated through catalytic hairpin assembly (CHA) facilitates intercalation of MB, which is then reduced to methylene white (MW) due to the electrons released from the GOD-mediated glucose oxidation. This process results in an amplified open circuit voltage (OCV) signal that can be monitored using a smartphone. The reduction in blue MB into colourless MW enables a colourimetric detection signal, which can also be quantified using a smartphone. Figures are adapted with permission from references [271,292], respectively.
Figure 7. Schematic illustration of EC nanobiosensor fabrication for miRNA sensing using single- (a) and dual-mode (EC/colourimetric) (b) detection. In single-mode sensor (a), a screen-printed carbon electrode (SPCE) modified with polypyrrole (PPY)-graphene nanocomposite, AuNP-conjugated ssDNA probe, and 6-mercapto-1-hexanol (MCH) acts as a sensor probe. When the target miRNA is present, methylene blue (MB) intercalates with the hybridised ssDNA probe-miRNA duplex, generating a strong peak current. In contrast, a significantly smaller peak current is observed in the absence of the target miRNA due to lesser intercalation of MB. The dual-mode sensor (b) employs carbon paper (CP) patterned with AuNP-vanadium sulphide nanosheet self-assembled nanorod (AuNP-VS4 NS NR) nanocomposite, modified with the H1 probe to function as the EC sensor probe. In parallel, the colourimetric sensor probe is created by modifying the AuNP-VS4 NS NR patterned CP with glucose oxidase (GOD). In the presence of target miRNA, a y-shaped DNA generated through catalytic hairpin assembly (CHA) facilitates intercalation of MB, which is then reduced to methylene white (MW) due to the electrons released from the GOD-mediated glucose oxidation. This process results in an amplified open circuit voltage (OCV) signal that can be monitored using a smartphone. The reduction in blue MB into colourless MW enables a colourimetric detection signal, which can also be quantified using a smartphone. Figures are adapted with permission from references [271,292], respectively.
Biosensors 15 00639 g007
Figure 8. Schematic illustration of fluorescence nanobiosensor fabrication for miRNA sensing using FRET-based approach. Fluorescently labelled DNA probes and graphene oxide (GO) (a) serve as donor-quencher pairs and are combined with the isothermal strand displacement amplification (iSDA) technique for the detection of target miRNAs. In the absence of target miRNA, no iSDA occurs, and the signals from all three probes, P1, P2, and P3, remain quenched by GO. When the target miRNA is present, it initiates the amplification process through iSDA, leading to the formation of P1-miRNA duplexes that prevent P1 from adsorbing onto the GO surface (signal-on). Meanwhile, P2 and P3 remain single-stranded and readily adsorb onto the GO surface. This causes their signals to be quenched by FRET (signal-off). In a label-free approach (b), silicon quantum dots (Si QDs) and gold nanoparticles (GNPs) were utilised as donor-acceptor pairs. In the absence of the miRNA target, FRET occurs (signal-off) due to the hybridisation between the triplex molecular beacon (tMB) and the triplex-forming oligonucleotide (TFO), bringing the Si QD and GNP into close proximity. However, when the miRNA target is present, the distance between the Si QD and GNP increases due to the hybridisation of tMB with the miRNA target, thereby restoring the Si QD fluorescence signal (signal-on). Figures are adapted with permission from references [276,295], respectively.
Figure 8. Schematic illustration of fluorescence nanobiosensor fabrication for miRNA sensing using FRET-based approach. Fluorescently labelled DNA probes and graphene oxide (GO) (a) serve as donor-quencher pairs and are combined with the isothermal strand displacement amplification (iSDA) technique for the detection of target miRNAs. In the absence of target miRNA, no iSDA occurs, and the signals from all three probes, P1, P2, and P3, remain quenched by GO. When the target miRNA is present, it initiates the amplification process through iSDA, leading to the formation of P1-miRNA duplexes that prevent P1 from adsorbing onto the GO surface (signal-on). Meanwhile, P2 and P3 remain single-stranded and readily adsorb onto the GO surface. This causes their signals to be quenched by FRET (signal-off). In a label-free approach (b), silicon quantum dots (Si QDs) and gold nanoparticles (GNPs) were utilised as donor-acceptor pairs. In the absence of the miRNA target, FRET occurs (signal-off) due to the hybridisation between the triplex molecular beacon (tMB) and the triplex-forming oligonucleotide (TFO), bringing the Si QD and GNP into close proximity. However, when the miRNA target is present, the distance between the Si QD and GNP increases due to the hybridisation of tMB with the miRNA target, thereby restoring the Si QD fluorescence signal (signal-on). Figures are adapted with permission from references [276,295], respectively.
Biosensors 15 00639 g008
Figure 9. Schematic illustration of CL nanobiosensor fabrication for the detection of miRNA. During the assay, a FAM-labelled hairpin loop (I-1), functionalised with an HRP-mimicking DNAzyme (hemin-intercalated G-quadruplex) serves as the chemiluminescence donor, while magnetic graphene oxide (MGO) functions as the acceptor. In the absence of the target miRNA, the HRP-mimicking DNAzyme/FAM DNA probe remains in a folded conformation, effectively masking the luminescence signal generated by luminol/H2O2 catalysis via chemiluminescence resonance energy transfer (CRET) (signal-off). However, when the target miRNA is present, the hairpin loop unfolds and hybridises with the target. This interaction increases the distance between the probe DNA and GO, thus preventing CRET (signal-on) and enabling detection of the target. The figure is reproduced with permission from the reference [298].
Figure 9. Schematic illustration of CL nanobiosensor fabrication for the detection of miRNA. During the assay, a FAM-labelled hairpin loop (I-1), functionalised with an HRP-mimicking DNAzyme (hemin-intercalated G-quadruplex) serves as the chemiluminescence donor, while magnetic graphene oxide (MGO) functions as the acceptor. In the absence of the target miRNA, the HRP-mimicking DNAzyme/FAM DNA probe remains in a folded conformation, effectively masking the luminescence signal generated by luminol/H2O2 catalysis via chemiluminescence resonance energy transfer (CRET) (signal-off). However, when the target miRNA is present, the hairpin loop unfolds and hybridises with the target. This interaction increases the distance between the probe DNA and GO, thus preventing CRET (signal-on) and enabling detection of the target. The figure is reproduced with permission from the reference [298].
Biosensors 15 00639 g009
Figure 10. Schematic illustration of colourimetric miRNA detection techniques using AuNP aggregation (a), HRP enzyme (b), and peroxidase-mimic nanozyme (c). In the aggregation approach (a), when the target miRNA is absent, the ssDNA probe-functionalized AuNPs remain dispersed, exhibiting a red colour due to the repulsion between negatively charged DNA molecules. However, in the presence of target miRNA, hybridisation occurs between the ssDNA probes and the target miRNA, bringing the AuNPs into close proximity and resulting in aggregation, which produces a blue/purple colour. In the enzyme-based approach (b), the presence of target miRNA facilitates the formation of a duplex between polystyrene particles and HRP-conjugated ssDNA probes (PS-HRP-conjugated ssDNA probe). This duplex is recognised by the DSN enzyme, which specifically cleaves the hybridised DNA, leaving the miRNA intact and freeing HRP to catalyse the oxidation of peroxidase substrates (TMB/H2O2), thereby generating visual signals. In the absence of the miRNA target, the DSN enzyme cannot recognise the PS-HRP-conjugated ssDNA probe, leading to HRP remaining bound and preventing the oxidation of the chromogenic substrate (TMB). In the nanozyme-based approach (c), when target miRNA is present, magnetic bead-conjugated DNA probe 1 (MB-conjugated-P1) forms a duplex and subsequently creates a sandwich structure with probe 2 (P2). Hairpins 1 (H1) and 2 (H2) initiate a hybridisation chain reaction (HCR), resulting in the formation of a super sandwich structure on the MBs. The addition of copper salt (Cu2+) and ascorbic acid (AA) leads to the generation of peroxidase-mimicking copper nanoparticles (CuNPs), which intercalate into the duplex structure and catalyse the oxidation of the TMB substrate to create a visual signal. Conversely, in the absence of target miRNA, the HCR cannot proceed, preventing Cu2+ from interacting with the H1/H2 DNA strands, thus hindering the formation of CuNPs and the subsequent generation of a visual signal. Figures are adapted with permission from references [288,303], and [302], respectively.
Figure 10. Schematic illustration of colourimetric miRNA detection techniques using AuNP aggregation (a), HRP enzyme (b), and peroxidase-mimic nanozyme (c). In the aggregation approach (a), when the target miRNA is absent, the ssDNA probe-functionalized AuNPs remain dispersed, exhibiting a red colour due to the repulsion between negatively charged DNA molecules. However, in the presence of target miRNA, hybridisation occurs between the ssDNA probes and the target miRNA, bringing the AuNPs into close proximity and resulting in aggregation, which produces a blue/purple colour. In the enzyme-based approach (b), the presence of target miRNA facilitates the formation of a duplex between polystyrene particles and HRP-conjugated ssDNA probes (PS-HRP-conjugated ssDNA probe). This duplex is recognised by the DSN enzyme, which specifically cleaves the hybridised DNA, leaving the miRNA intact and freeing HRP to catalyse the oxidation of peroxidase substrates (TMB/H2O2), thereby generating visual signals. In the absence of the miRNA target, the DSN enzyme cannot recognise the PS-HRP-conjugated ssDNA probe, leading to HRP remaining bound and preventing the oxidation of the chromogenic substrate (TMB). In the nanozyme-based approach (c), when target miRNA is present, magnetic bead-conjugated DNA probe 1 (MB-conjugated-P1) forms a duplex and subsequently creates a sandwich structure with probe 2 (P2). Hairpins 1 (H1) and 2 (H2) initiate a hybridisation chain reaction (HCR), resulting in the formation of a super sandwich structure on the MBs. The addition of copper salt (Cu2+) and ascorbic acid (AA) leads to the generation of peroxidase-mimicking copper nanoparticles (CuNPs), which intercalate into the duplex structure and catalyse the oxidation of the TMB substrate to create a visual signal. Conversely, in the absence of target miRNA, the HCR cannot proceed, preventing Cu2+ from interacting with the H1/H2 DNA strands, thus hindering the formation of CuNPs and the subsequent generation of a visual signal. Figures are adapted with permission from references [288,303], and [302], respectively.
Biosensors 15 00639 g010
Figure 11. Schematic illustration of the fabrication of EC antibody- (a) and aptamer-based (b) nanobiosensor for leptin detection. In antibody-based biosensor (a), a reduced graphene oxide (rGO)-Au nanocomposite patterned electrode is modified with bionanocapsule-ZZ (ZZ-BNC) and bovine serum albumin (BSA). The rGO-Au patterning ensures signal amplification, while ZZ-BNC facilitates the vertical orientation of antibodies, and BSA prevents non-specific interactions at the electrode surface. In the presence of leptin, the binding between leptin and the antibody hinders efficient electron transfer due to increase in charge transfer resistance, thereby causing a decrease in the EC signal. Conversely, in the absence of leptin, electron transfer occurs efficiently, resulting in a high EC signal. In the aptamer-based biosensor (b), a pair of aptamers is employed to create a sandwich-type biosensor. Zinc oxide nanoparticles (ZnO NPs), AuNPs, and capture aptamers (Apt 1) are utilised to modify the screen-printed electrode (SPE), while alkaline phosphatase (ALP)-labelled aptamers (Apt 2) serve as detection aptamers. When leptin is present, a sandwich complex forms that traps leptin between the capture and detection aptamers. This results in ALP converting the inactive α-naphthyl phosphate substrate into the electroactive product α-naphthol, producing a strong current signal. In the absence of leptin, however, this sandwich complex does not form on the electrode surface, inhibiting the generation of the electroactive product and resulting in weak current signals. Figures are adapted with permission from references [319,325], respectively.
Figure 11. Schematic illustration of the fabrication of EC antibody- (a) and aptamer-based (b) nanobiosensor for leptin detection. In antibody-based biosensor (a), a reduced graphene oxide (rGO)-Au nanocomposite patterned electrode is modified with bionanocapsule-ZZ (ZZ-BNC) and bovine serum albumin (BSA). The rGO-Au patterning ensures signal amplification, while ZZ-BNC facilitates the vertical orientation of antibodies, and BSA prevents non-specific interactions at the electrode surface. In the presence of leptin, the binding between leptin and the antibody hinders efficient electron transfer due to increase in charge transfer resistance, thereby causing a decrease in the EC signal. Conversely, in the absence of leptin, electron transfer occurs efficiently, resulting in a high EC signal. In the aptamer-based biosensor (b), a pair of aptamers is employed to create a sandwich-type biosensor. Zinc oxide nanoparticles (ZnO NPs), AuNPs, and capture aptamers (Apt 1) are utilised to modify the screen-printed electrode (SPE), while alkaline phosphatase (ALP)-labelled aptamers (Apt 2) serve as detection aptamers. When leptin is present, a sandwich complex forms that traps leptin between the capture and detection aptamers. This results in ALP converting the inactive α-naphthyl phosphate substrate into the electroactive product α-naphthol, producing a strong current signal. In the absence of leptin, however, this sandwich complex does not form on the electrode surface, inhibiting the generation of the electroactive product and resulting in weak current signals. Figures are adapted with permission from references [319,325], respectively.
Biosensors 15 00639 g011
Figure 12. Schematic illustration of colourimetric nanobiosensor fabrication for RBP-4 (a) and IL-6 (b) detection. For RBP-4 detection (a), aptamer-functionalised AuNPs are utilised as sensor probes. In the presence of RBP-4, the aptamers desorb from the AuNP surface to interact with the target, leaving the AuNP surface unprotected against salt-induced aggregation (purple colour). Conversely, in the absence of RBP-4, the aptamers remain attached to the AuNPs, protecting them from aggregation and, hence, exhibiting a red colour. For IL-6 detection (b), a sandwich-type ELISA was developed by incorporating anti-IL-6 as the capture antibody and peroxidase-mimicking Pd@Pt core–shell nanodendrites-modified anti-IL-6 as the detection antibody. In the presence of IL-6, a sandwich complex is formed, trapping IL-6 between the capture and detection antibodies. This leads to peroxidase nanozyme-mediated TMB/H2O2 catalysis, resulting in a visual signal that correlates to the IL-6 concentration. In contrast, when the target is absent, the sandwich complex does not form, preventing the detection antibody from binding to the microtiter plate. As a result, TMB/H2O2 catalysis is inhibited, and no visual signal is produced. Figures are adapted with permission from references [312,333], respectively.
Figure 12. Schematic illustration of colourimetric nanobiosensor fabrication for RBP-4 (a) and IL-6 (b) detection. For RBP-4 detection (a), aptamer-functionalised AuNPs are utilised as sensor probes. In the presence of RBP-4, the aptamers desorb from the AuNP surface to interact with the target, leaving the AuNP surface unprotected against salt-induced aggregation (purple colour). Conversely, in the absence of RBP-4, the aptamers remain attached to the AuNPs, protecting them from aggregation and, hence, exhibiting a red colour. For IL-6 detection (b), a sandwich-type ELISA was developed by incorporating anti-IL-6 as the capture antibody and peroxidase-mimicking Pd@Pt core–shell nanodendrites-modified anti-IL-6 as the detection antibody. In the presence of IL-6, a sandwich complex is formed, trapping IL-6 between the capture and detection antibodies. This leads to peroxidase nanozyme-mediated TMB/H2O2 catalysis, resulting in a visual signal that correlates to the IL-6 concentration. In contrast, when the target is absent, the sandwich complex does not form, preventing the detection antibody from binding to the microtiter plate. As a result, TMB/H2O2 catalysis is inhibited, and no visual signal is produced. Figures are adapted with permission from references [312,333], respectively.
Biosensors 15 00639 g012
Figure 13. Schematic illustration of fluorescence-based nanobiosensor fabrication for vaspin (a) and IL-6 (b) detection. Vaspin detection is mediated on a sandwich-format lateral flow device (LFD) by utilising UCNPs as fluorescence labels and aptamers as MREs. The UCNP-aptamer conjugates are pre-immobilised on the conjugate pad. In the presence of vaspin, these conjugates form a UCNP-aptamer-vaspin complex, which is then captured by biotin-labelled primary aptamers at the test line, creating a sandwich structure. Unbound or excess UCNP-aptamer conjugates are captured by biotin-labelled secondary aptamers at the control line. Fluorescence signals at both the test and control lines indicate the presence of vaspin, while a lack of signal at the test line confirms its absence. IL-6 detection (b) was facilitated using FRET on/off mechanism, wherein nitrogen-doped carbon quantum dots (NCQ) act as donors and aptamer-conjugated AuNPs as acceptors. In the absence of IL-6 or the presence of non-target adipokine, the NCQ is able to interact with aptamers via π-π binding and their fluorescence is quenched by AuNPs (FRET on). However, in the presence of IL-6, the aptamers bind to the target IL-6, preventing NCQ adsorption and allowing their fluorescence to persist (FRET off), with the intensity corresponding to the concentration of IL-6. Figures are adapted with permission from references [328,332], respectively.
Figure 13. Schematic illustration of fluorescence-based nanobiosensor fabrication for vaspin (a) and IL-6 (b) detection. Vaspin detection is mediated on a sandwich-format lateral flow device (LFD) by utilising UCNPs as fluorescence labels and aptamers as MREs. The UCNP-aptamer conjugates are pre-immobilised on the conjugate pad. In the presence of vaspin, these conjugates form a UCNP-aptamer-vaspin complex, which is then captured by biotin-labelled primary aptamers at the test line, creating a sandwich structure. Unbound or excess UCNP-aptamer conjugates are captured by biotin-labelled secondary aptamers at the control line. Fluorescence signals at both the test and control lines indicate the presence of vaspin, while a lack of signal at the test line confirms its absence. IL-6 detection (b) was facilitated using FRET on/off mechanism, wherein nitrogen-doped carbon quantum dots (NCQ) act as donors and aptamer-conjugated AuNPs as acceptors. In the absence of IL-6 or the presence of non-target adipokine, the NCQ is able to interact with aptamers via π-π binding and their fluorescence is quenched by AuNPs (FRET on). However, in the presence of IL-6, the aptamers bind to the target IL-6, preventing NCQ adsorption and allowing their fluorescence to persist (FRET off), with the intensity corresponding to the concentration of IL-6. Figures are adapted with permission from references [328,332], respectively.
Biosensors 15 00639 g013
Table 1. Diagnostic criteria for diabetes and prediabetes.
Table 1. Diagnostic criteria for diabetes and prediabetes.
ConditionADA Criteria [37]WHO Criteria [44]
NormalFPG ≤ 100 mg/dL (≤5.5 mmol/L)
2 h PG during OGTT ≤ 140 mg/dL (≤7.7 mmol/L)
HbA1c ≤ 5.7%
DiabetesFPG ≥ 126 mg/dL (≥7.0 mmol/L)
RBG ≥ 200 mg/dL (≥11.1 mmol/L)
2 h PG during OGTT ≥ 200 mg/dL (≥11.1 mmol/L)
HbA1c ≥ 6.5%
Pre-diabetesFPG 101–124 mg/dL
(5.6–6.9 mmol/L)
FPG 110–124 mg/dL
(6.1–6.9 mmol/L)
HbA1c 5.7–6.4%HbA1c 6.0–6.4%
2 h PG during OGTT 140–198 mg/dL (7.8–11.0 mmol/L)
Table 3. Emerging biomarkers in T2D.
Table 3. Emerging biomarkers in T2D.
Biomarker TypeExamplesRole in T2DReference
Glucose metabolism biomarkersHbA1c, fasting glucose, glycated albumin, fructosamine, 1,5-AHGMarkers of impaired glucose metabolism and progression to T2D.[140,145,146,147,148]
Insulin-Related biomarkersFasting insulin, C-peptide, HOMA-IR, proinsulinIndicators of insulin resistance and beta-cell dysfunction.[149,150,151,152,153]
MetabolitesBCAAs, acylcarnitines, tyrosine, phenylalanine, glycineIndicators of altered metabolic pathways related to glucose and lipid metabolism.[154,155,156,157,158]
Lipid biomarkersTriglycerides, HDL, LDL, free fatty acids, lipoprotein(a) Markers of dyslipidaemia and metabolic syndrome.[159,160,161,162,163]
Genetic biomarkersTCF7L2, FTO, PPARG, KCNJ11, SLC30A8, CDKN2A/B, TOX, HHEX, IGF2BP2Variants in genes associated with β-cell function, insulin sensitivity, and glucose metabolism.[159,160,161,162,163,164,165,166]
Inflammatory biomarkersCRP, IL-6, TNF-α, IL-1β, MCP-1Indicators of chronic inflammation linked to insulin resistance.[167,168,169,170,171,172]
Gut MicrobiotaReduced Akkermansia muciniphila, increased Firmicutes/Bacteroidetes ratio, reduced butyrate levelsPatterns of gut microbiota composition linked to glucose metabolism and inflammation.[173,174,175,176,177]
Proteomic biomarkersFetuin-A, ceruloplasmin, FGF21, SHBGIndicators of protein-related pathways influencing glucose metabolism and insulin resistance.[178,179,180,181]
Table 4. The classification of adipokines and their association with T2D.
Table 4. The classification of adipokines and their association with T2D.
ClassificationAdipokineFunctionAssociation with T2DReference(s)
Anti-inflammatoryAdiponectinEnhances insulin sensitivity, reduces inflammation, and promotes fatty acid oxidation.Reduced levels are associated with insulin resistance and T2D.[186,187]
VaspinInhibits pro-inflammatory cytokines, enhances insulin sensitivity, and regulates metabolic homeostasis.Elevated levels correlate with obesity and impaired insulin sensitivity[188,189]
Omentin-1Improves insulin sensitivity, modulates glucose metabolism, and suppresses inflammation.Reduced levels correlate with insulin resistance and inflammation.[190]
Gremlin-1Improves insulin sensitivity, promotes metabolic homeostasis; emerging therapeutic target.Increased levels correlate with obesity and insulin resistance.[191]
FGF-21Regulates glucose and lipid metabolism, reduces inflammation, and protects against metabolic stress.Increased levels are linked to metabolic dysfunctions in T2D.[192,193]
LeptinRegulates appetite and energy balance; elevated levels linked to chronic inflammation in obesity.Increased levels are associated with obesity and leptin resistance.[194]
Pro-inflammatoryResistinPromotes insulin resistance, increases pro-inflammatory cytokine production, and links obesity to T2D.Elevated levels are associated with an increased risk of T2D.[195,196]
ChemerinIncreases inflammation, regulates adipocyte differentiation, and promotes insulin resistance.Elevated levels are linked with metabolic syndrome and T2D.[197,198]
VisfatinEnhances inflammatory responses and may modulate insulin secretion.Elevated levels are observed in metabolic syndrome and T2D.[199]
RBP-4Induces insulin resistance, links adipose dysfunction to glucose intolerance, and promotes inflammation.Elevated levels are strongly associated with insulin resistance.[200]
TNF-αPromotes systemic inflammation and contributes to insulin resistance in obesity and T2D.High levels are linked with systemic inflammation and insulin resistance[201]
IL-6Induces chronic low-grade inflammation, impairs insulin signalling and metabolic regulation.Elevated levels (>3 pg/mL) correlate with systemic inflammation and T2D.[202]
Table 5. Nanobiosensor-based detection of miRNAs under different disease conditions 1.
Table 5. Nanobiosensor-based detection of miRNAs under different disease conditions 1.
miRNAHealth ConditionMode of Detection and TechniqueNanomaterial(s)Analytical PerformanceRemarksReference
miR-21Lung cancerEC using OCV/
colourimetric dual-mode
VS4 NS-NR-AuNP hybrid nanocompositeLR = 0.001–103 pg/mL
LOD = 0.16 fg/mL
Recovery = 84.1–100.6%
Required amplification = no
Real sample validation = yes
[271]
miR-21CancerEC using OCV/
colourimetric dual-mode
Graphdiyne/AuNPs compositesLR = 0.5 fM–100 pM
LOD = 0.15 fM (EC)
33 fM (Colourimetric)
Recovery = 97.6–105.7%
Required amplification = yes
Real sample validation = yes
[272]
miR-141Breast cancerEC using OCV/
colourimetric dual-mode
Graphdiyne/AuNPs compositesLR = 0.0001–100 pM
LOD = 21.9 aM
Recovery = 99.0–106.3%
Required amplification = no
Real sample validation = yes
[273]
miR-141CancerECL/SPC
dual-mode
S-BN QDs and
WO3-x nanodots
LR = 10−17–10−10 M
LOD = 10−17 M
Recovery = 99.0–103%
Required amplification = no
Real sample validation = yes
[274]
miR-21Ovarian cancerFluorescenceAgNCsLR = 9 pM–1.55 nM
LOD = 2 pM
Recovery = 93–108%
Required amplification = no
Real sample validation = yes
[275]
miR-155CancerFluorescenceSi QDs and AuNPsLR = 10–800 pM
LOD = 10 pM
Recovery = 99–100.2%
Required amplification = no
Real sample validation = yes
[276]
miR-155CancerFluorescenceMSN@R6G
@AuNP
LR = 100 fM–100 nM
LOD = 2.18 fM
Recovery = 99–100.2%
Required amplification = no
Real sample validation = yes
[277]
miR-155CancerFluorescenceLa(III)-MOF and AgNPsLR = 2.7 fM–0.01 pM
LOD = 5.2 fM
Recovery = NR
Required amplification = no
Clinical sample validation = yes
[278]
miR-125a-5pAcute ischemic strokeFluorescenceAuNPsLR = 0.01–10 µM
LOD = NR
Recovery = 100.6–107.4%
Required amplification = yes
Real sample validation = yes
[279]
miR-222CancerFluorescenceUCNPs LR = 0.5–2.5 nM
LOD = 0.077 nM
Recovery = 97.6–102.1%
Required amplification = no
Real sample validation = yes
[280]
miR-362Inflammatory bowel disease FluorescenceAgNCsLR = 20–200 nM
LOD = 6.5 nM
Recovery = 74.7–92.8%
Required amplification = no
Real sample validation = yes
[281]
miR-499Acute
myocardial infarction
FluorescenceMoS2 NSLR = 0.1–13.33 nM
LOD = 381.8 pM
Recovery = 89.5–97.6%
Required amplification = yes
Real sample validation = yes
[282]
miR-21 and miR-155CancerFluorescence QDsLR = 0.1 pM–0.01 nM (both miRNA)
LOD = 0.1 pM
(both miRNA)
Recovery = NR
Required amplification = no
Clinical sample validation = yes
No labelling required
[283]
miR-21 and miR-155Breast and lung cancerFluorescence/
photothermal dual-mode
CeO2@AuNPLR = 10 pM–100 nM (miR-21) and
10 pM–200 nM
(miR-155)
LOD = 7.2 pM (miR-21) and 9.1 pM (miR-155)
Recovery = NR
Required amplification = yes
Clinical sample validation = yes
Nanozyme-based detection
[284]
miR-21 and miR-155CancerFluorescence (FL)/CL dual-mode
AuNPsLR = 500 fM–10 nM (FL)
10 fM–10 nM (CL) for both miRNAs
LOD = 107.2 fM (FL), 8.3 fM (CL) for miR-21 and 97.7 fM (FL), 11.2 fM (CL) for miR-155
Recovery = 93.4–121.8% (FL)
87.4–110.6% (CL) for miR-155
Required amplification = yes
Real sample validation = yes
[285]
miR-21CancerColourimetricAg/PtNCsLR = 10–1000 pM
LOD = 4.1 pM
Recovery = 93.8–106%
Required amplification = no
Real sample validation = yes
Nanozyme-based detection on µPAD
[286]
miR-21CancerColourimetricFe3O4 NPsLR = 5 fM–10 pM
LOD = 5 fM
Recovery = 97.5 –104%
Required amplification = yes
Real sample validation = yes
[287]
miR-155CancerColourimetric CuNPs LR = 80–500 aM
LOD = 22 aM
Recovery = 98.8–103.4%
Required amplification = yes
Real sample validation = yes
Nanozyme-based detection
[288]
miR-155CancerColourimetricAuNPsLR = 1–100 nM
LOD = 0.7 nM
Recovery = 96.8–103%
Required amplification = no
Real sample validation = yes
[289]
miR-148a Gastric cancerColourimetricAuNPsLR = NR
LOD = 1.9 nM
Recovery = NR
Required amplification = no
Real sample validation = no
[290]
1 This table is not exhaustive and only compiles studies published in the last five years, 2019–2024.
Table 6. Nanobiosensor-based adipokine detection under different disease conditions 1.
Table 6. Nanobiosensor-based adipokine detection under different disease conditions 1.
AdipokineHealth Condition(s)Mode of Detection and TechniqueNanomaterialAnalytical PerformanceRemarksReference
LeptinObesityEC using EIS FeNi NPsLR = 0.5 pg/mL–80 ng/mL
LOD = 157.4 fg/mL
Recovery = NR
Antibody-based detection.
Real sample validation matrix was mouse serum.
[322]
LeptinNeuro-degenerative disease and brain trauma EC using DPV ZnO NPs and AuNPsLR = 0.01–100 pg/mL
LOD = 0.035 pg/mL
Recovery = 96.3–103.4% (serum)
97.8–108.8% (plasma)
Aptamer pair-based detection.
Real sample validation matrix was serum and plasma.
[319]
LeptinTraumatic
brain injury
EC using EIS TiO2 NPs and AuNPsLR = 1–100 pg/mL and
100–1000 pg/mL
LOD = 0.31 pg/mL
Recovery = 96.4–107.8%
Aptamer-based detection.
Real sample validation matrix was serum and plasma.
[321]
LeptinObesity-related conditionsEC using DPV rGO and AuNPsLR = 0.001–1000 pg/mL
LOD = 0.87 fg/mL
Recovery = NR
Antibody-based detection.
Real sample validation matrix was mice and human serum
[325]
LeptinNon-alcoholic fatty liver EC using SWVPorous graphene functionalised black phosphorous and AuNPsLR = 0.15 pg/mL–2.5 ng/mL
LOD = 0.036 fg/mL
Recovery = 98–100.8%
Antibody-based detection.
Real sample validation matrix was serum.
[326]
LeptinObesityEC using DPVCe3NbO7/CeO2 hollow nanospheresLR = 0.5 pg/mL–
12 ng/mL
LOD = 0.14 fg/mL
Recovery = 86.05–94.7%
Antibody-based detection.
Real sample validation matrix was plasma
[330]
TNF-α and IL-6Inflammation and immune responseEC using chronoamperometry (CA)rGO and AuNPsLR = 0.1–500 pg/mL
LOD = 0.69 pg/mL
Recovery = NR
Antibody-based detection. Real samples were not tested.[329]
IL-6Colorectal cancerEC using EISAuNPsLR = 5 pg/mL–10 ng/mL
LOD = 1.6 pg/mL
Recovery = 103.9–109.6%
Aptamer-based detection.
Real sample validation matrix was serum.
[331]
TNF-αInflammatory and autoimmune diseasesFluorescenceZnO NRsLR = 0.1–100 fg/mL
LOD = NR
Recovery = NR
Antibody-based detection. Real samples were not tested.[318]
VaspinObesity and T2DFluorescenceUCNPs LR = 0.1–55 ng/mL
LOD = 39 pg/mL
Recovery = 96.9–101%
Aptamer pair-based detection on an LFD.
Real sample validation matrix was serum.
[328]
IL-6Infectious diseaseFluorescenceCDs and AuNPsLR = 1.5–5.9 pg/mL
LOD = 0.82 pg/mL
Recovery = 95.7–102.9%
Aptamer-based detection.
Real sample validation matrix was serum.
[332]
IL-6 Inflammatory diseasesColourimetricPd@Pt NDs LR = 0.05–2 pg/mL
LOD = 0.04 pg/mL
Recovery = 93.7–105.5%
Nanozyme-based detection using antibodies.
Real sample validation matrix was serum.
[333]
VisfatinMetabolic disorderColourimetricPtNP-modified Ce-MOFLR = 1–100 ng/mL
LOD = 0.11
pg/mL
Recovery = 96.7–98.6%
Nanozyme-based detection using aptamer.
Real sample validation matrix was serum.
[327]
RBP-4Obesity and T2DColourimetricAuNPsLR = 0–125 nM
LOD = 90.76 nM
Recovery = NR
Salt-induced NP aggregation mediated aptamer-based detection.
Real sample validation was not performed.
[312]
IL-6InflammationColourimetricAuNPsLR = NR
LOD = 1.95 µg/mL
Recovery = NR
Salt-induced NP aggregation-based detection by using aptamer-pair.
Real samples were not tested.
[314]
IL-6Infectious diseaseColourimetricAuNPsLR = 1 fg/mL–100 pg/mL
LOD = 1 fg/mL
Recovery = NR
Antibody-mediated detection on paper-based analytical device.
Real sample validation matrix was human blood and bronchial aspirate.
[334]
IL-6Cardiovascular disordersColourimetricMagnetic beadsLR = 1.6–100 ng/mL
LOD = 0.4 ng/mL
Recovery = 99–114%
Antibody-based microfluidic detection.
Real sample validation matrix was saliva.
[335]
TNF-α and IL-6Immuno-modulation therapySPRAuNRsLR = NR
LOD = 69.15 pg/mL (TNF-α)
0.93 pg/mL (IL-6) Recovery = NR
Multiplex detection using nano-plasmonic ink and antibodies.
Practical validation was performed in an immunomodulated macrophage sample.
[316]
RBP-4Acute kidney injurySPRMNPsLR = NR
LOD = 84 pg/mL
Recovery = 96.7–98.6%
Smartphone-assisted antibody-mediated detection.
Real sample validation matrix was urine.
[320]
IL-6Cancer, cardiovascular disorders, diabetes, rheumatoid arthritisSERSAu@Ag-Au core–shell LR = 100 fg/mL–
1 ng/mL
LOD = 12.4 fg/mL
Recovery = 92.4–105.3%
Antibody-based detection. Real sample validation matrix was spiked serum saliva and clinical serum samples. [324]
AdiponectinGDMSERSAuNTs LR = 0.1 ng/mL–1 µg/mL
LOD = 30 fg/mL
Recovery = 93.1–106.02%
Antibody-mediated detection.
Real sample validation matrix was serum from pregnant women
[315]
RBP-4Obesity and T2DCLAuNPsLR = 0.001–2 ng/mL
LOD = 1 pg/mL
Recovery = NR
Aptamer-based detection.
Practical application was validated using patient samples.
[313]
1 This table is not exhaustive and only compiles studies from the last five years, 2019–2024.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Baranwal, A.; Bansal, V.; Shukla, R. Emerging Biomarkers and Nanobiosensing Strategies in Diabetes. Biosensors 2025, 15, 639. https://doi.org/10.3390/bios15100639

AMA Style

Baranwal A, Bansal V, Shukla R. Emerging Biomarkers and Nanobiosensing Strategies in Diabetes. Biosensors. 2025; 15(10):639. https://doi.org/10.3390/bios15100639

Chicago/Turabian Style

Baranwal, Anupriya, Vipul Bansal, and Ravi Shukla. 2025. "Emerging Biomarkers and Nanobiosensing Strategies in Diabetes" Biosensors 15, no. 10: 639. https://doi.org/10.3390/bios15100639

APA Style

Baranwal, A., Bansal, V., & Shukla, R. (2025). Emerging Biomarkers and Nanobiosensing Strategies in Diabetes. Biosensors, 15(10), 639. https://doi.org/10.3390/bios15100639

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop