Next Article in Journal
Chemical Composition, Nutritional, and Biological Properties of Extracts Obtained with Different Techniques from Aronia melanocarpa Berries
Previous Article in Journal
Total Syntheses and Antibacterial Studies of Natural Isoflavones: Scandenone, Osajin, and 6,8-Diprenylgenistein
Previous Article in Special Issue
Inhibition of MMP-2 and MMP-9 by Dietary Antioxidants in THP-1 Macrophages and Sera from Patients with Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Role of Phenolic Compounds in Chronic Kidney Disease: A Systematic Review

by
Filipa Baptista
1,*,
Jessica Paié-Ribeiro
2,
Mariana Almeida
2 and
Ana Novo Barros
1,*
1
Centre for the Research and Technology of Agro-Environmental and Biological Sciences, CITAB, University of Trás-os-Montes and Alto Douro, UTAD, 5000-801 Vila Real, Portugal
2
CECAV—Animal and Veterinary Research Centre, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
*
Authors to whom correspondence should be addressed.
Molecules 2024, 29(11), 2576; https://doi.org/10.3390/molecules29112576
Submission received: 15 April 2024 / Revised: 20 May 2024 / Accepted: 30 May 2024 / Published: 30 May 2024

Abstract

:
Chronic kidney disease (CKD) presents a formidable global health concern, affecting one in six adults over 25. This review explores the potential of phenolic compounds in managing CKD and its complications. By examining the existing research, we highlight their diverse biological activities and potential to combat CKD-related issues. We analyze the nutritional benefits, bioavailability, and safety profile of these compounds. While the clinical evidence is promising, preclinical studies offer valuable insights into underlying mechanisms, optimal dosages, and potential side effects. Further research is crucial to validate the therapeutic efficacy of phenolic compounds for CKD. We advocate for continued exploration of their innovative applications in food, pharmaceuticals, and nutraceuticals. This review aims to catalyze the scientific community’s efforts to leverage phenolic compounds against CKD-related challenges.

1. Introduction

Chronic kidney disease (CKD) is a widely prevalent and grave issue that has a detrimental impact on human health, curtails lifespan, and escalates healthcare expenditures globally [1]. Chronic kidney disease has become a significant public health concern worldwide due to its increasing incidence and prevalence in both developed and developing nations. Notably, the kidneys show a more significant age-associated chronic pathology compared to other organs, such as the brain, liver, and heart. According to recent studies, it has been found that a substantial percentage of the adult population above the age of 25, precisely one in six individuals, is affected by CKD to some extent. Moreover, the incidence of CKD is known to increase with age, which has resulted in a staggering 1.2 million deaths and 28.0 million years of lives lost annually [2,3]. It is noteworthy that this disease is projected to become the fifth leading cause of death worldwide by 2040, manifesting as one of the largest expected increases in any major cause of death [4].
Studies have shown notable disparities in CKD prevalence among various racial and ethnic populations. For example, African Americans, Hispanic Americans, and Indigenous populations tend to have higher rates of CKD compared to Caucasians [5]. These disparities are influenced by a multitude of factors, including genetic predisposition, socioeconomic status, access to healthcare, and cultural practices [5,6].
Furthermore, CKD demonstrates distinct patterns in terms of sex-based differences. While both men and women are susceptible to CKD, there are variations in its prevalence and disease progression between the sexes. While men may exhibit a slightly higher prevalence of CKD in certain populations, women often experience faster progression to end-stage renal disease (ESRD) once diagnosed with CKD [7,8]. These sex-specific differences underscore the importance of considering gender-related factors in CKD research and management strategies.
There are many causes and consequences of chronic kidney disease, including those that are common and well researched, such as diabetes, obesity, and cardiovascular disease, as well as complications due to oxidative stress, as presented in Figure 1. However, causation in chronic kidney disease is not yet fully understood. The structural features of CKD include increased interstitial fibrosis, tubular atrophy, renal vasculopathy, glomerulosclerosis, and reduced renal regenerative capability. Atherosclerosis, an arterial disease, is very prevalent in CKD patients and is the leading cause of cardiovascular disease. It is mainly characterized by low levels of high-density lipoproteins (HDLs) and increased plasma triglycerides because of very low-density lipoprotein (LDL) accumulation. Oxidative stress and inflammation aid in the development and progression of atherosclerosis and consequently of CKD. In recent years, several reports have shown indications of a rise in oxidative stress among patients undergoing renal replacement therapy and hemodialysis. However, the importance of increased oxidative stress in renal patients needs further clarification through clinical endpoint studies. This is especially crucial since studies on antioxidant treatments to prevent vascular and other diseases in non-renal patients have produced ambiguous results.
In 2015, United Nations member countries rallied around a common objective: the Sustainable Development Goals (SDGs). A pivotal health-related target within these goals is a reduction in mortality due to non-communicable diseases (NCDs) by one-third by 2030 [5]. Chronic kidney disease (CKD) has emerged as a significant global public health challenge, evidenced by its escalating incidence and prevalence rates, which have surged by approximately 5 percent over the last four years [6].
Hence, it becomes imperative to delve into the pathophysiological mechanisms underlying CKD to unearth novel therapeutic avenues capable of either preventing or decelerating the progression of this disease.
Over the past decade, there has been a remarkable increase in interest among both consumers and the scientific community regarding phenolic compounds and their antioxidant activity. This upsurge can be attributed to epidemiological studies that have established a clear link between the consumption of diets enriched with natural antioxidants and a reduced risk of diseases associated with oxidative stress, including but not limited to cancer and cardiovascular disease [9].
In the current review, we will discuss the explored effects of phenolic compounds on the treatment and progression of CKD and related diseases.

1.1. Phenolic Compounds

Phenolic compounds, a diverse group of plant-derived phytochemicals, encompass various classes, each with unique chemical structures and biological activities. These compounds play crucial roles in plant defense mechanisms and contribute significantly to human health due to their antioxidant, anti-inflammatory, and other bioactive properties [10,11,12].
Flavonoids, vital secondary metabolites within plants, exhibit a widespread presence across a plethora of botanical sources, including fruits, vegetables, herbs, stems, cereals, nuts, flowers, and seeds [13,14,15]. With over 10,000 distinct compounds identified to date, flavonoids showcase a remarkable diversity [16,17]. Their manifold biochemical and antioxidant properties render them potent agents in combating various ailments, such as cardiovascular disease, cancer, and neurodegenerative disorders [18,19,20,21]. Moreover, flavonoids contribute to a broad spectrum of health benefits and serve as indispensable constituents in nutraceuticals, pharmaceuticals, and medicinal and cosmetic formulations [22]. This multifaceted utility primarily stems from their anti-inflammatory, antioxidant, anticarcinogenic, and antimutagenic attributes, alongside their capacity to modulate crucial cellular enzyme functions [23,24].
Flavonoids exhibit remarkable structural diversity, leading to their classification into various types based on their chemical structure, degree of unsaturation, and carbon ring oxidation. These include flavones, flavanones, isoflavones, flavonols, chalcones, flavanols, and anthocyanins, each of which enjoys wide distribution throughout nature [25]. Phenolic acids, another prominent class, are categorized into hydroxybenzoic acids (e.g., gallic acid) and hydroxycinnamic acids (e.g., caffeic acid). Widely distributed in fruits, vegetables, whole grains, and coffee, phenolic acids possess antioxidant, antimicrobial, and anti-inflammatory properties, contributing to their potential health benefits [26,27].
Tannins, characterized by their ability to bind and precipitate proteins, are subdivided into hydrolyzable tannins (e.g., ellagitannins) and condensed tannins (proanthocyanidins). Found in foods such as grapes, nuts, and tea, tannins exhibit antioxidant, antimicrobial, and anti-inflammatory activities and are associated with various health benefits [28,29].
Lignans, abundant in flaxseeds, sesame seeds, and whole grains, are phytoestrogens known for their potential hormone-balancing effects. These compounds possess antioxidant and anti-inflammatory properties and have been studied for their potential role in reducing the risk of hormone-related cancers and cardiovascular diseases [30,31].
Stilbenes, such as resveratrol, found in grapes and red wine, are known for their cardioprotective and anti-aging effects. Resveratrol in particular has garnered significant attention for its antioxidant, anti-inflammatory, and potential anticancer properties [32,33].

1.2. Phenolic Compounds and Their Potential Therapeutic Benefits in Chronic Kidney Diease

Phenolic compounds have garnered attention for their potential therapeutic benefits in chronic kidney disease (CKD) [34,35]. These compounds encompass a diverse array of phytochemicals, including flavonoids, phenolic acids, tannins, and lignans, known for their antioxidant and anti-inflammatory properties [36,37].
In the context of CKD, where oxidative stress and inflammation play pivotal roles in disease progression, phenolic compounds offer promise as natural agents capable of mitigating these pathological processes [38,39,40]. Their ability to scavenge free radicals and modulate inflammatory pathways makes them attractive candidates for adjunctive CKD therapy [40,41,42].
Research suggests that phenolic compounds may exert renoprotective effects according to several mechanisms [34,43,44]. Firstly, their antioxidant properties help counteract the oxidative stress burden imposed on renal tissues in CKD, thereby preserving renal function and attenuating disease progression. Additionally, phenolic compounds possess anti-inflammatory properties, which can mitigate the inflammatory cascade implicated in CKD pathogenesis [34,42,45].
Moreover, phenolic compounds exhibit potential in ameliorating other CKD-associated complications, such as cardiovascular disease, by modulating lipid metabolism, improving endothelial function, and reducing arterial stiffness [38,41,46].
While much of the evidence supporting the beneficial effects of phenolic compounds in CKD is derived from preclinical studies and epidemiological observations, ongoing research endeavors aim to elucidate their specific mechanisms of action and therapeutic potential in human subjects [47].
In summary, phenolic compounds represent a promising avenue in the quest for novel CKD therapies, offering the potential to mitigate oxidative stress, inflammation, and associated complications. Further research is warranted to elucidate their precise mechanisms of action and establish their efficacy and safety profiles in CKD management [47,48].

2. Results

A flow diagram summarizing the literature search process, screening, and the selection of potential studies is depicted in Figure 2. Initially, 654 papers were retrieved through keyword searches and Boolean operators. Following the application of exclusion criteria within the databases, the number of studies was reduced by 72%, resulting in a total of 183 scientific articles. From this pool, 71 articles were ultimately selected for further analysis. Studies briefly mentioning chronic kidney disease without clinical studies were excluded.
The analysis revealed a sharp rise in publication numbers, particularly prominent in 2020 and 2022, with Brazil emerging as the most prolific contributor. Notably, a significant portion of the literature pertaining to herbal medicine and chronic kidney disease was centred around rat models. In the Discussion section, we elucidate the primary findings extracted from our comprehensive literature review. These results are systematically categorized into thematic clusters to enhance the clarity of the current research landscape. Furthermore, we embark on an exploration of the broader implications of these discoveries, underscoring their pivotal role in advancing our understanding of physiological mechanisms and potential therapeutic avenues.

3. Discussion

Chronic kidney disease (CKD) is a growing health concern worldwide. While treatment options exist, managing CKD often involves a multi-pronged approach, including dietary modifications. Research hints at the potential benefits of dietary interventions in slowing the progression of CKD and improving the overall health outcomes for patients. For instance, the study by Choi et al. [49] concluded that individuals who consumed a plant-based diet, rich in polyphenols, were less likely to experience a deterioration of kidney function, especially among participants with initial-stage CKD characterized as mild. Additionally, extra virgin olive oil (EVOO), also rich in phenolics, might improve kidney function and reduce cardiovascular risks in CKD patients, as suggested by Romani et al. [50] and Marrone et al. [51]. While Silva et al. [52] found limited evidence for EVOO’s impact on specific CKD biomarkers in healthy individuals, the research on CKD patients offers promise. Jespersen et al. [53,54] determined a possible association between moderate wine consumption and a lower prevalence of CKD. Currently, Anvarifard et al. [55] is assessing the effectiveness of propolis supplementation in 44 eligible CKD patients through a multi-centred, randomized, double-blind, placebo-controlled clinical trial. If the results of this study reveal the remarkable effectiveness of propolis in improving the quality of life and clinical outcomes in patients with CKD, this compound may reach a new milestone as an adjunctive therapy for CKD, and it opens a new window for further studies.
By exploring these studies, we aim to shed light on potential dietary strategies that, alongside traditional medical management, can contribute to a more comprehensive approach to CKD management. Nonetheless, it is critical to understand CKD stands as a complex health challenge, exacerbated by various contributing factors.

3.1. The Role of Oxidative Stress in Chronic Kidney Disease Pathogenesis

Oxidative stress plays a pivotal role in the pathogenesis of chronic kidney disease (CKD), highlighting the imbalance between reactive oxygen species (ROS) and antioxidant defenses, which initiates a cascade of renal tissue damage [14,15]. This stress can inflict damage on DNA, exacerbating conditions such as CKD.
The nuclear factor (erythroid-derived 2)-like factor 2 (Nrf2) pathway serves as a defense mechanism against oxidative stress, as illustrated in Figure 3 [56]. Under normal conditions, Nrf2 is sequestered in the cytoplasm by an inhibitory protein known as Kelch-like ECH-associated protein 1 (Keap1). However, when cells encounter oxidative stress, Keap1 releases Nrf2, allowing it to translocate to the nucleus. Once inside the nucleus, Nrf2 binds to specific DNA sequences known as antioxidant response elements (AREs), facilitating the expression of various antioxidant and detoxification enzymes [16]. These enzymes play a crucial role in neutralizing ROS and safeguarding cells from oxidative damage.
The activation of Nrf2 by oxidative stress indirectly inhibits nuclear factor kappa B (NF-κB) by upregulating the expression of antioxidant enzymes that scavenge ROS, thereby mitigating the stimuli for NF-κB activation [17]. Upon activation, NF-κB initiates the expression of genes involved in inflammation, including cytokines and adhesion molecules [18]. While these inflammatory responses can be helpful in the short term for fighting infections, chronic activation of NF-κB can contribute to tissue damage and worsen existing conditions like CKD. Additionally, factors like transforming growth factor beta (TGF-β) and tumor necrosis factor alpha (TNF-α) can also influence NF-κB activity. When TNF-α binds to its receptor, it triggers a signaling cascade that leads to NF-κB activation and promotes inflammatory responses [57]. In some cell types, TGF-β can actually suppress NF-κB activity, potentially mitigating inflammation. However, in other contexts, TGF-β can also activate NF-κB, contributing to processes like fibrosis observed in CKD [58]. Polyphenols like curcumin [57], epigallocatechin-3-gallate (EGCG) [58], resveratrol [59], a synthetic caffeamide derivative [60], and lignophenol [61] are believed to exert their beneficial effects by modulating these pathways, as seen for polyphenol-rich foods and drinks, such as dark chocolate [62,63], Clitoria ternatea flower petal extract [64,65], açaí [66,67], mushrooms [68,69], corn silk [70], sorghum [71], probiotic drinks [72], and unfermented grape juice [73]. They can activate Nrf2, promoting the antioxidant defense system, and/or inhibit NF-κB, TGF-β, and/or TNF-α, thereby reducing inflammation and the associated oxidative stress in CKD patients [74]. Indeed, the interactions between these pathways are intricate and remain the subject of ongoing research. Nevertheless, this simplified explanation offers a foundational understanding of how these pathways intersect and how polyphenols may potentially intervene in this interplay to mitigate the burden of oxidative stress in CKD.

3.2. Diabetes and Dietary Sugar Consumption

Uncontrolled diabetes leads to chronically elevated blood sugar (hyperglycemia). This hallmark feature directly damages the delicate filtration units (glomeruli) within the kidneys. Initially, the kidneys attempt to compensate for this stress by working harder, leading to an increased glomerular filtration rate (GFR). However, this hyperfiltration further strains the kidneys over time. Additionally, hyperglycemia promotes the leakage of protein (albumin) into the urine (proteinuria), an early indicator of kidney damage [75,76]. It should not be neglected how high blood sugar triggers inflammatory responses and accelerates the formation of scar tissue (fibrosis) within the kidneys, ultimately leading to impaired kidney function, while also considering the local variations in the mechanical properties [77] at specific organ regions, which can lead to different liver states in both health and disease [78]. Excessive sugar intake, particularly refined sugars and sugar-sweetened beverages, significantly contributes to hyperglycemia. This dietary habit not only increases the risk of developing type 2 diabetes but also potentiates CKD in diabetic individuals [76]. Furthermore, high sugar consumption often coincides with a diet high in saturated fats and low in fiber, further promoting metabolic syndrome, a cluster of conditions including obesity, insulin resistance, and high blood pressure, all of which further increase the risk of CKD. While all sugars can contribute to diabetes and CKD, fructose (abundant in fruits and added to processed foods) may have a more detrimental effect on kidney health [75]. This is because fructose is primarily metabolized in the liver, where it can further contribute to dyslipidemia and oxidative stress [76]. Additionally, individual susceptibility to the effects of sugar on blood sugar and kidney function can vary. Regular monitoring of blood sugar and kidney function through urine or blood tests is crucial for early detection and intervention to prevent or slow CKD progression. Certain polyphenols might improve insulin sensitivity, leading to better blood sugar control and reducing the workload on the kidneys [79,80,81]; modulate enzymes involved in carbohydrate metabolism, potentially helping to regulate blood sugar levels [82,83]; and may reduce the risk factors for type 2 diabetes by protecting against glucolipotoxicity-induced damage [84], helping prevent diabetic nephropathy [85,86,87,88,89].
By understanding the intricate link between diabetes, sugar consumption, and CKD, we can better understand the role polyphenols play in mitigating diabetic nephropathy and CKD.

3.3. Hypertension and Cardiovascular Disease

Hypertension and cardiovascular disease (CVD) can significantly exacerbate chronic kidney disease (CKD) through a vicious cycle. Elevated blood pressure places strain on the delicate filtration system within the kidneys, leading to gradual damage to the glomeruli (microscopic filters) and tubules, ultimately resulting in diminished kidney function and the progression of CKD [90]. Furthermore, uncontrolled hypertension constricts blood vessels throughout the body, including those that supply the kidneys. This reduced blood flow restricts the delivery of oxygen and nutrients to the kidneys, further compromising their function.
CVD often entails chronic inflammation throughout the body [91], which can harm the kidneys by promoting scarring and a decline in kidney function. Atherosclerosis, characterized by plaque build-up in the arteries due to CVD, can also impact the arteries supplying the kidneys. This narrowing of the blood vessels due to atherosclerosis mimics the effects of hypertension, exacerbating CKD. Additionally, when the heart weakens due to CVD, it struggles to effectively pump blood, leading to fluid build-up in the body, including around the kidneys, which can exacerbate kidney damage.
Taken together, hypertension and CVD hasten the decline in kidney function [91]. The impaired waste removal ability of the kidneys further elevates blood pressure, perpetuating a detrimental cycle. This accelerated progression of CKD heightens the risk of complications such as end-stage renal disease (ESRD) and necessitates dialysis [91].
Research suggests that polyphenols may offer protective effects against CVD by enhancing blood vessel function, thereby potentially improving blood pressure [92,93,94,95,96,97,98]. They may also inhibit LDL oxidation, a pivotal step in arterial plaque formation, and reduce inflammation [99,100,101]. While evidence of the role of polyphenols in ESRD is still emerging and not as robust as that for CVD and hypertension, preliminary studies on the consumption of green tea [102,103] and coffee [104] show promising outcomes.

3.4. Nephrotoxicity

Nephrotoxicity, characterized by a rapid decline in kidney function due to exposure to toxins or medications, can present in diverse forms. Understanding these distinct injury patterns is essential for accurate diagnosis, effective treatment, and potentially preventing further damage. Nephrotoxicity arises from disruptions in the blood flow to the kidneys, leading to a reduced glomerular filtration rate (GFR), resulting in diminished urine output and elevated blood urea nitrogen (BUN) and creatinine levels [105]. Various toxins and medications, including antibiotics, like aminoglycosides (gentamicin, tobramycin) and certain cephalosporins (cephalexin), cisplatin, and other chemotherapeutic drugs commonly used for cancer treatment, as well as environmental toxins like heavy metals (cadmium, lead, mercury) and certain industrial chemicals, can instigate this type of injury, accumulating in the kidneys and damaging the tubules and surrounding tissue [105,106]. Additionally, although less prevalent, nephrotoxicity can occur due to crystal formation within the tubules, obstructing urine flow and inducing inflammation.
Research indicates that polyphenols may offer protective effects against nephrotoxicity. For instance, elderflower [107] and fish oil [108] significantly enhanced renal activity in gentamicin-induced nephrotoxicity in rat models, while bee propolis [109,110], fig leaves [111,112], avocado, walnuts [113,114], pitaya juice [115], black soybeans [116], and other medicinal plants [113,117,118,119] may reverse the renal damage caused by anticancer agents such as adriamycin, 5-fluorouracil, and cisplatin. Furthermore, guava leaves [120], stevia residue [121,122], and ginger [123] have demonstrated efficacy in attenuating medication-induced renal atrophy.
Further research has shown that polyphenol-rich foods, including Indian plums [124], radishes [125,126], green algae [127], acacia gum [128], basil leaves [129,130], essential oils [131], coffee and olive oils [132], and red palm oil [133,134], can ameliorate kidney damage induced by various toxins. Moreover, phenolic compounds have shown promising results in preventing further heavy-metal-induced kidney injury [135,136,137]. While research suggests that some polyphenols might hold promise in safeguarding the kidneys from nephrotoxic damage, further studies are warranted to confirm their efficacy and safety in human subjects.

3.5. Other Related Conditions

Beyond the previously discussed conditions, CKD can also be associated with hyperuremia (high blood uric acid) and urolithiasis (kidney stones). Hyperuricemia is common in chronic kidney disease (CKD) and may be present in 50% of patients presenting for dialysis, and while it is a risk factor for CKD, it is not entirely clear whether lowering uric acid levels can prevent or slow down CKD progression in all cases. Nonetheless, research shows the polyphenols present in both moringa leaves [138] and peony flowers [139,140] can effectively reduce the serum uric acid levels in rats by regulating serum xanthine oxidase activity and renal urate transporters. In addition, polyphenol-rich foods have shown diuretic properties that could help mitigate urolithiasis [141,142,143,144,145].

3.6. Polyphenol-Rich Foods and Their Reported Activity

Phenolic compounds are a diverse group of secondary metabolites found in plants, characterized by their aromatic ring structures with one or more hydroxyl groups attached. The structural diversity of phenolic compounds contributes to their wide range of bioactivities, which are determined by specific structural features [47,146].
For example, the number and position of hydroxyl groups on the aromatic ring influence the antioxidant activity of phenolic compounds. Compounds with more hydroxyl groups tend to exhibit higher antioxidant potential due to their ability to donate hydrogen atoms and scavenge free radicals, thus protecting cells from oxidative damage [147,148]. This structural configuration facilitates hydrogen atom donation, effectively neutralizing free radicals and protecting cells from oxidative damage. The Free Oxygen Radical Test (FORT) and total antioxidant capacity (TAC) assays serve as valuable tools for quantifying this enhanced antioxidant capacity [149,150].
Similarly, the presence of conjugated double bonds in the aromatic ring system enhances the anti-inflammatory properties of phenolic compounds. This structural feature allows phenolic compounds to modulate inflammatory pathways by inhibiting the production of pro-inflammatory mediators and cytokines like interleukin-6 (IL-6) [47,151]. By mitigating the activity of these inflammatory signaling molecules, phenolics can demonstrably decrease the levels of C-reactive protein (CRP) and the erythrocyte sedimentation rate (ESR)—established biomarkers of systemic inflammation [152]. Superoxide dismutase (SOD), an enzymatic defense mechanism against free radicals, may be indirectly influenced by certain phenolic structures. However, their primary impact is likely exerted on malondialdehyde (MDA), a byproduct of lipid peroxidation—a process triggered by free radicals. By effectively scavenging free radicals, phenolics can significantly reduce MDA levels, signifying decreased oxidative damage [153].
Additionally, the presence of certain functional groups, such as methoxy or glycoside groups, can affect the solubility and bioavailability of phenolic compounds, thereby influencing their biological activity. These structural modifications may also impact the interactions of phenolic compounds with enzymes, receptors, and other cellular targets, further contributing to their diverse bioactivities [154,155].
Overall, the structure of phenolic compounds plays a crucial role in determining their bioactivity, making them promising candidates for various applications in medicine, nutrition, and agriculture. Understanding the structure–function relationships of phenolic compounds can help in the design and development of novel therapeutic agents with enhanced efficacy and safety profiles [156,157].
Found abundantly in fruits, vegetables, grains, and beverages like tea and wine, these compounds are known for their antioxidant and anti-inflammatory properties. Their presence in foods underscores the importance of a varied and colorful diet for overall health and well-being. An overview of the phenolic compounds and their reported bioactivity in the studies included in this systematic review is presented in Table 1.

3.7. Notes on the Excluded Research

This review aimed to comprehensively evaluate the current research on the use of polyphenols and polyphenol-rich foods to treat CKD and CKD-related conditions. A rigorous selection process was employed to ensure the included studies met specific criteria related to the real impact on CKD. While a total of 183 studies were identified through our search strategy, 112 studies were ultimately excluded from this review.
This section details the rationale behind the exclusion of these studies. Understanding the limitations of the included research is crucial for interpreting the findings and identifying potential areas for future investigation. The excluded studies may have been focused on conditions or assessments not directly associated with CKD, with methodological limitations impacting the relevance of the results to this review, or may have lacked a control group.
The review focused on exploring the potential benefits of phenolic compounds in chronic kidney disease (CKD) and related conditions. Consequently, studies investigating the adverse health effects of specific phenolic compounds, such as p-Cresol, were deliberately excluded. p-Cresol, a methylated phenolic compound and gut microbiome metabolite, has been associated with documented negative effects on kidney function. While phenolic compounds encompass a diverse group with several health benefits, certain compounds like p-Cresol can have detrimental effects. It is crucial to note that p-Cresol is not typically derived from plants. Although many phenolic compounds are sourced from plant-based origins, p-Cresol primarily originates from two main sources. Firstly, it is produced through industrial processes, conventionally extracted from coal tar, a by-product of coal roasting for coke production. Secondly, p-Cresol is generated within the gut microbiome, through the bacterial fermentation of proteins in the large intestine. Given these origins and its established negative health impacts on the kidneys, p-Cresol was deemed irrelevant for the purposes of exploring the benefits of plant-based phenolics for CKD.
By presenting these considerations, our aim is to provide a transparent overview of the existing evidence and to identify areas where further research is needed to better understand the potential benefits of plant-based phenolics in CKD management.

4. Materials and Methods

This study employed a systematic selection of literature within the field of management research. The study’s overarching aims were exploratory and descriptive in nature, facilitating a comprehensive evaluation of the data and the acquisition of interpretative insights. The technical procedures employed in this research closely adhered to the systematic literature review framework, following the well-established steps of PRISMA. These steps encompassed formulating the research question (I), identifying pertinent databases (II), devising search strategies and selecting and accessing pertinent literature (III), evaluating the quality of the studies included (IV), and ultimately conducting an in-depth analysis, synthesis, and dissemination of the research findings (V).

4.1. Formulatation of the Research Question (I)

Research Question: What is the scientific research scenario on the use of phenolic compounds in the prevention, treatment, and management of chronic kidney disease?

4.2. Searched Databases and the Search Strategies (II)

Conducting a systematic review depends on the scope and quality of the included studies. Thus, to recover scientific articles of proven quality, we sought to use only electronic bases that retrieve journals that perform peer reviews of a manuscript, which were national and international. The bases used were Web of Science, Scopus, PubMed, and ScienceDirect. Next, each database’s descriptors and the Booleans used as search guidance were determined. The search method varied several times among databases because they have some peculiarities. Boolean operators were used to establish the selection criteria and thus retrieve the maximum number of papers related to the theme. All bases were last consulted on the 11th of February. The descriptors used as search guidance in each database were determined. Keywords were established according to the selection criteria, and thus the maximum number of papers related to the theme was retrieved. When searching, the AND operator was inserted between two words to obtain articles that contain both (e.g., “phenolic compounds” AND “chronic kidney disease”). Keywords between quotation marks were used to refine the search. Therefore, articles were retrieved only if the words appeared together. In all the databases, no time/date limit was imposed. After obtaining the results, only scientific articles were selected, excluding books and book chapters, review articles, conference papers, notes, letters, errata, and editorials.
The searches in the Scopus, PubMed, Web of Science, and ScienceDirect databases were similar. The following keywords and combinations were used through Booleans to refine the search and exclude irrelevant articles. The search was not restricted (All fields) in the Web of Science database and restricted to the title, abstract, and keywords in all the other databases to the following combination: “phenolic compounds” AND “chronic kidney disease”. After obtaining the results, only scientific articles were selected, excluding books and book chapters, review articles, notes, letters, conference papers, errata, and editorials. Duplicates in the same database were discarded.

4.3. Inclusion and Exclusion Criteria (III)

The articles retrieved from the databases were selected according to the following criteria:
Only studies where phenolic compounds were used to prevent, treat, or manage CDK and CKD-related conditions were selected. After being selected according to the mentioned criteria, the articles were grouped into different knowledge fields: Oxidative stress and signaling pathways, diabetes and dietary sugar consumption, hypertension and cardiovascular disease, nephrotoxicity, and other related conditions. Records were marked as ineligible when they only briefly mentioned CKD and made no significant remarks on how the study impacted this disease, namely studies that only studied the phytochemical profile of food matrices.

4.4. Critical Analysis of the Selected Studies (IV)

The articles chosen in the previous step were read in full to extract relevant aspects of the objectives, methodology, results, and conclusions. This analysis also considered the quality of the studies, which required a detailed description of the methodology and conclusive results with a thorough discussion.

4.5. Summary of the Results (V)

The presentation of the results focused on describing the main characteristics of the studies, highlighting the effects of phenolic compounds on CKD, either in preventing, treating, or managing the disease.

5. Conclusions

Chronic kidney disease (CKD) is a global health issue affecting around 850 million individuals worldwide, particularly in low- and middle-income nations. Dietary polyphenols have emerged as a promising avenue for the treatment and management of CKD. Recent studies have shed light on the mechanisms through which polyphenols can alleviate CKD-related conditions. However, despite valuable insights from the existing research, several unanswered questions and knowledge gaps persist.
Exploring potential avenues for future research is essential to advance our understanding of how CKD affects different organs. This includes investigating the long-term effects of dietary interventions, delving into the underlying mechanisms of the observed benefits, and conducting larger and more diverse human clinical trials. Addressing these research gaps is pivotal in developing more effective strategies for managing and potentially preventing CKD.
Polyphenols and other food phenolics have garnered increasing interest in the scientific community due to their potential to promote health and well-being. However, more research is needed to understand the metabolism and availability of these compounds in the body, essential factors for unlocking their beneficial effects on human and animal health. Future research should focus on exploring new cultivars to identify novel compounds and elucidate their mechanisms for medical and pharmaceutical purposes. Phytochemical characterization is indispensable to driving progress in discovering and developing new drugs based on natural compounds.
Recent research suggests that polyphenols may hold significant promise in CKD care, offering potential advantages such as slowing disease progression, enhancing kidney function, and mitigating the cardiovascular risks associated with CKD. However, the successful integration of polyphenols into CKD treatment necessitates patient education and active participation to ensure adherence to dietary changes.
This review highlights the importance of a collaborative, multidisciplinary approach involving nephrologists, dietitians, and other healthcare professionals to delivering comprehensive CKD care. Synthesizing diverse research findings not only advances our understanding of CKD but also serves as a practical guide for clinicians and researchers navigating the complexities of CKD treatment and management.
Ultimately, this synthesis contributes to the ongoing discourse on refining strategies to enhance the quality of life for CKD patients worldwide, underscoring the promising role of dietary polyphenols in this endeavor. Therefore, in-depth research into exploiting new cultivars, understanding the mechanisms of action, and enhancing accessibility to CKD treatments is imperative for fostering a more sustainable future and promoting global health and well-being.

Author Contributions

Conceptualization, A.N.B. and F.B.; methodology, F.B.; validation, A.N.B.; formal analysis, F.B.; data curation, F.B., J.P.-R. and M.A.; writing—original draft preparation, F.B., M.A. and J.P.-R.; writing—review and editing, A.N.B., F.B., M.A. and J.P.-R.; supervision, A.N.B.; funding acquisition, A.N.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by national funds from FCT—Portuguese Foundation for Science and Technology under the project UIDB/04033/2020 (https://doi.org/10.54499/UIDB/04033/2020, https://doi.org/10.54499/LA/P/0126/2020 (accessed on 4 March 2024)).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Small, D.M.; Coombes, J.S.; Bennett, N.; Johnson, D.W.; Gobe, G.C. Oxidative Stress, Anti-Oxidant Therapies and Chronic Kidney Disease. Nephrology 2012, 17, 311–321. [Google Scholar] [CrossRef] [PubMed]
  2. Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.; Abebe, M.; Adebayo, O.M.; Afarideh, M.; Agarwal, S.K.; Agudelo-Botero, M.; et al. Global, Regional, and National Burden of Chronic Kidney Disease, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [PubMed]
  3. Xie, Y.; Bowe, B.; Mokdad, A.H.; Xian, H.; Yan, Y.; Li, T.; Maddukuri, G.; Tsai, C.Y.; Floyd, T.; Al-Aly, Z. Analysis of the Global Burden of Disease Study Highlights the Global, Regional, and National Trends of Chronic Kidney Disease Epidemiology from 1990 to 2016. Kidney Int. 2018, 94, 567–581. [Google Scholar] [CrossRef] [PubMed]
  4. Foreman, K.J.; Marquez, N.; Dolgert, A.; Fukutaki, K.; Fullman, N.; McGaughey, M.; Pletcher, M.A.; Smith, A.E.; Tang, K.; Yuan, C.W.; et al. Forecasting Life Expectancy, Years of Life Lost, and All-Cause and Cause-Specific Mortality for 250 Causes of Death: Reference and Alternative Scenarios for 2016–40 for 195 Countries and Territories. Lancet 2018, 392, 2052–2090. [Google Scholar] [CrossRef] [PubMed]
  5. Lopes, A.A. Relationships of Race and Ethnicity to Progression of Kidney Dysfunction and Clinical Outcomes in Patients with Chronic Kidney Failure. Adv. Ren. Replace. Ther. 2004, 11, 14–23. [Google Scholar] [CrossRef] [PubMed]
  6. Brown, J.S.; Elliott, R.W. Social Determinants of Health: Understanding the Basics and Their Impact on Chronic Kidney Disease. Nephrol. Nurs. J. 2021, 48, 131–145. [Google Scholar] [CrossRef] [PubMed]
  7. Carrero, J.J.; Hecking, M.; Chesnaye, N.C.; Jager, K.J. Sex and Gender Disparities in the Epidemiology and Outcomes of Chronic Kidney Disease. Nat. Rev. Nephrol. 2018, 14, 151–164. [Google Scholar] [CrossRef]
  8. García, G.G.; Iyengar, A.; Kaze, F.; Kierans, C.; Padilla-Altamira, C.; Luyckx, V.A. Sex and Gender Differences in Chronic Kidney Disease and Access to Care around the Globe. Semin. Nephrol. 2022, 42, 101–113. [Google Scholar] [CrossRef] [PubMed]
  9. Troncoso, N.; Sierra, H.; Carvajal, L.; Delpiano, P.; Günther, G. Fast High Performance Liquid Chromatography and Ultraviolet-Visible Quantification of Principal Phenolic Antioxidants in Fresh Rosemary. J. Chromatogr. A 2005, 1100, 20–25. [Google Scholar] [CrossRef]
  10. Liu, W.; Cui, X.; Zhong, Y.; Ma, R.; Liu, B.; Xia, Y. Phenolic Metabolites as Therapeutic in Inflammation and Neoplasms: Molecular Pathways Explaining Their Efficacy. Pharmacol. Res. 2023, 193, 106812. [Google Scholar] [CrossRef]
  11. Sun, W.; Shahrajabian, M.H. Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health. Molecules 2023, 28, 1845. [Google Scholar] [CrossRef] [PubMed]
  12. Nurzyńska-Wierdak, R. Phenolic Compounds from New Natural Sources—Plant Genotype and Ontogenetic Variation. Molecules 2023, 28, 1731. [Google Scholar] [CrossRef]
  13. Kumar, L.; Bharti; Goutam, E.; Choudhary, D. Perspectives of Secondary Metabolites in Reference to Vegetable Crops: A Review. Int. J. Environ. Clim. Chang. 2023, 13, 873–882. [Google Scholar] [CrossRef]
  14. Gupta, M.; Mishra, A. Bioactive Flavonoids: A Comparative Overview of the Biogenetic and Chemical Synthesis Approach. Mini-Rev. Med. Chem. 2023, 23, 1818–1837. [Google Scholar] [CrossRef]
  15. Dave Mehta, S.; Upadhyay, S.; Rai, G. Importance of Flavonoid as Secondary Metabolites. In Flavonoid Metabolism—Recent Advances and Applications in Crop Breeding; Intechopen: London, UK, 2023. [Google Scholar] [CrossRef]
  16. Nabil-Adam, A.; Elnosary, M.E.; Ashour, M.L.; Abd El-Moneam, N.M.; Shreadah, M.A. Flavonoids Biosynthesis in Plants as a Defense Mechanism: Role and Function Concerning Pharmacodynamics and Pharmacokinetic Properties. In Flavonoid Metabolism—Recent Advances and Applications in Crop Breeding; Intechopen: London, UK, 2023. [Google Scholar] [CrossRef]
  17. Safe, S.; Jayaraman, A.; Chapkin, R.S.; Howard, M.; Mohankumar, K.; Shrestha, R. Flavonoids: Structure–Function and Mechanisms of Action and Opportunities for Drug Development. Toxicol. Res. 2021, 37, 147–162. [Google Scholar] [CrossRef]
  18. Ravindran, R.; Swamy, M.K.; Jaganathan, R. Therapeutic Potential of Plant Polyphenolics and Their Mechanistic Action against Various Diseases. Nat. Bio-Act. Compd. Chem. Pharmacol. Health Care Pract. 2019, 2, 313–351. [Google Scholar] [CrossRef] [PubMed]
  19. Shoaib, S.; Ansari, M.A.; Al Fatease, A.; Safhi, A.Y.; Hani, U.; Jahan, R.; Alomary, M.N.; Ansari, M.N.; Ahmed, N.; Wahab, S.; et al. Plant-Derived Bioactive Compounds in the Management of Neurodegenerative Disorders: Challenges, Future Directions and Molecular Mechanisms Involved in Neuroprotection. Pharmaceutics 2023, 15, 749. [Google Scholar] [CrossRef]
  20. Khan, J.; Deb, P.K.; Priya, S.; Medina, K.D.; Devi, R.; Walode, S.G.; Rudrapal, M. Dietary Flavonoids: Cardioprotective Potential with Antioxidant Effects and Their Pharmacokinetic, Toxicological and Therapeutic Concerns. Molecules 2021, 26, 4021. [Google Scholar] [CrossRef] [PubMed]
  21. Hernández-Rodríguez, P.; Baquero, L.P.; Larrota, H.R. Flavonoids. In Bioactive Compounds—Health Benefits and Potential Applications; Woodhead Publishing: Cambridge, UK, 2019; pp. 265–288. [Google Scholar] [CrossRef]
  22. Khan, A.K.; Kousar, S.; Tungmunnithum, D.; Hano, C.; Abbasi, B.H.; Anjum, S. Nano-Elicitation as an Effective and Emerging Strategy for in Vitro Production of Industrially Important Flavonoids. Appl. Sci. 2021, 11, 1694. [Google Scholar] [CrossRef]
  23. Lo, A.H.; Liang, Y.C.; Lin-Shiau, S.Y.; Ho, C.T.; Lin, J.K. Carnosol, an Antioxidant in Rosemary, Suppresses Inducible Nitric Oxide Synthase through down-Regulating Nuclear Factor-ΚB in Mouse Macrophages. Carcinogenesis 2002, 23, 983–991. [Google Scholar] [CrossRef]
  24. Kang, H.; Kim, B. Bioactive Compounds as Inhibitors of Inflammation, Oxidative Stress and Metabolic Dysfunctions via Regulation of Cellular Redox Balance and Histone Acetylation State. Foods 2023, 12, 925. [Google Scholar] [CrossRef] [PubMed]
  25. Koes, R.E.; Quattrocchio, F.; Mol, J.N.M. The Flavonoid Biosynthetic Pathway in Plants: Function and Evolution. BioEssays 1994, 16, 123–132. [Google Scholar] [CrossRef]
  26. Rashmi, H.B.; Negi, P.S. Phenolic Acids from Vegetables: A Review on Processing Stability and Health Benefits. Food Res. Int. 2020, 136, 109298. [Google Scholar] [CrossRef] [PubMed]
  27. Ji, L.; Deng, H.; Xue, H.; Wang, J.; Hong, K.; Gao, Y.; Kang, X.; Fan, G.; Huang, W.; Zhan, J.; et al. Research Progress Regarding the Effect and Mechanism of Dietary Phenolic Acids for Improving Nonalcoholic Fatty Liver Disease via Gut Microbiota. Compr. Rev. Food Sci. Food Saf. 2023, 22, 1128–1147. [Google Scholar] [CrossRef] [PubMed]
  28. Farha, A.K.; Yang, Q.Q.; Kim, G.; Li, H.B.; Zhu, F.; Liu, H.Y.; Gan, R.Y.; Corke, H. Tannins as an Alternative to Antibiotics. Food Biosci. 2020, 38, 100751. [Google Scholar] [CrossRef]
  29. Tong, Z.; He, W.; Fan, X.; Guo, A. Biological Function of Plant Tannin and Its Application in Animal Health. Front. Vet. Sci. 2022, 8, 803657. [Google Scholar] [CrossRef] [PubMed]
  30. Jang, W.Y.; Kim, M.Y.; Cho, J.Y. Antioxidant, Anti-Inflammatory, Anti-Menopausal, and Anti-Cancer Effects of Lignans and Their Metabolites. Int. J. Mol. Sci. 2022, 23, 15482. [Google Scholar] [CrossRef] [PubMed]
  31. Soleymani, S.; Habtemariam, S.; Rahimi, R.; Nabavi, S.M. The What and Who of Dietary Lignans in Human Health: Special Focus on Prooxidant and Antioxidant Effects. Trends Food Sci. Technol. 2020, 106, 382–390. [Google Scholar] [CrossRef]
  32. Navarro-Orcajada, S.; Conesa, I.; Vidal-Sánchez, F.J.; Matencio, A.; Albaladejo-Maricó, L.; García-Carmona, F.; López-Nicolás, J.M. Stilbenes: Characterization, Bioactivity, Encapsulation and Structural Modifications. A Review of Their Current Limitations and Promising Approaches. Crit. Rev. Food Sci. Nutr. 2023, 63, 7269–7287. [Google Scholar] [CrossRef]
  33. Vijayan, N.; Haridas, M.; Abdulhameed, S. Stilbenes and Their Derivatives in Traditional Medicine. Bioresour. Bioprocess Biotechnol. 2017, 2, 407–418. [Google Scholar] [CrossRef]
  34. Hefer, M.; Huskic, I.M.; Petrovic, A.; Raguz-Lucic, N.; Kizivat, T.; Gjoni, D.; Horvatic, E.; Udiljak, Z.; Smolic, R.; Vcev, A.; et al. A Mechanistic Insight into Beneficial Effects of Polyphenols in the Prevention and Treatment of Nephrolithiasis: Evidence from Recent In Vitro Studies. Crystals 2023, 13, 1070. [Google Scholar] [CrossRef]
  35. Pandey, K.B.; Rizvi, S.I. Plant Polyphenols as Dietary Antioxidants in Human Health and Disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
  36. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [PubMed]
  37. Fraga, C.G.; Croft, K.D.; Kennedy, D.O.; Tomás-Barberán, F.A. The Effects of Polyphenols and Other Bioactives on Human Health. Food Funct. 2019, 10, 514–528. [Google Scholar] [CrossRef] [PubMed]
  38. Vagkopoulou, A.; Haddad, N.; Kontogiorgos, I.; Papatolios, T.; Papadopoulou, E.; Makridis, D.; Tsimikas, S.; Kalaitzidis, R.; Karasavvidou, D. #3813 Effect of Oral Flavonoids on Arterial Stiffness in Ckd: A Pilot Prospective Study. Nephrol. Dial. Transplant. 2023, 38, gfad063d_3813. [Google Scholar] [CrossRef]
  39. Haddad, N.; Bagopoulou, N.; Kontogiorgos, I.; Papatolios, T.; Papadopoulou, E.; Makridis, D.; Tzimikas, S.; Fountoglou, A.; Kalaitzidis, R.; Karasavvidou, D. Effect of Oral Flavonoids on Arterial Stiffness in Ckd—A Pilot Prospective Study. J. Hypertens. 2023, 41, e262. [Google Scholar] [CrossRef]
  40. Supriyadi, R.; Koswara, M.I.A.; Soelaeman, M.A.; Huang, I. The Effect of Antioxidants Supplementation on Oxidative Stress and Proinflammatory Biomarkers in Patients with Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Eur. Rev. Med. Pharmacol. Sci. 2023, 27, 1413–1426. [Google Scholar] [CrossRef] [PubMed]
  41. Avila-Carrasco, L.; García-Mayorga, E.A.; Díaz-Avila, D.L.; Garza-Veloz, I.; Martinez-Fierro, M.L.; González-Mateo, G.T. Potential Therapeutic Effects of Natural Plant Compounds in Kidney Disease. Molecules 2021, 26, 6096. [Google Scholar] [CrossRef]
  42. Nayak, S.S.; Bhaijamal, R.A.; Vaghasia, J.; Shivani, R. Role of Plant Based Natural Antioxidents on Chronic Kidney Disease. Natl. J. Pharm. Sci. 2023, 3, 101–106. [Google Scholar] [CrossRef]
  43. Casanova, A.G.; López-Hernández, F.J.; Vicente-Vicente, L.; Morales, A.I. Are Antioxidants Useful in Preventing the Progression of Chronic Kidney Disease? Antioxidants 2021, 10, 1669. [Google Scholar] [CrossRef]
  44. Chtourou, Y.; Morjen, M.; Ammar, R.; Mhiri, R.; Jemaà, M.; ELBini-Dhouib, I.; Fetoui, H.; Srairi-Abid, N.; Marrakchi, N.; Jebali, J. Investigation of the Renal Protective Effect of Combined Dietary Polyphenols in Streptozotocin-Induced Diabetic Aged Rats. Nutrients 2022, 14, 2867. [Google Scholar] [CrossRef] [PubMed]
  45. Alsawaf, S.; Alnuaimi, F.; Afzal, S.; Thomas, R.M.; Chelakkot, A.L.; Ramadan, W.S.; Hodeify, R.; Matar, R.; Merheb, M.; Siddiqui, S.S.; et al. Plant Flavonoids on Oxidative Stress-Mediated Kidney Inflammation. Biology 2022, 11, 1717. [Google Scholar] [CrossRef] [PubMed]
  46. Elgadir, M.A.; Chigurupati, S.; Mariod, A.A. Selected Potential Pharmaceutical and Medical Benefits of Phenolic Compounds: Recent Advances. Funct. Food Sci. 2023, 3, 108–128. [Google Scholar] [CrossRef]
  47. Rahman, M.M.; Rahaman, M.S.; Islam, M.R.; Rahman, F.; Mithi, F.M.; Alqahtani, T.; Almikhlafi, M.A.; Alghamdi, S.Q.; Alruwaili, A.S.; Hossain, M.S.; et al. Role of Phenolic Compounds in Human Disease: Current Knowledge and Future Prospects. Molecules 2022, 27, 233. [Google Scholar] [CrossRef] [PubMed]
  48. Olson, K.R.; Derry, P.J.; Kent, T.A.; Straub, K.D. The Effects of Antioxidant Nutraceuticals on Cellular Sulfur Metabolism and Signaling. Antioxid. Redox Signal. 2023, 38, 68–94. [Google Scholar] [CrossRef] [PubMed]
  49. Choi, Y.; Steffen, L.M.; Chu, H.; Duprez, D.A.; Gallaher, D.D.; Shikany, J.M.; Schreiner, P.J.; Shroff, G.R.; Jacobs, D.R. A Plant-Centered Diet and Markers of Early Chronic Kidney Disease during Young to Middle Adulthood: Findings from the Coronary Artery Risk Development in Young Adults (CARDIA) Cohort. J. Nutr. 2021, 151, 2721–2730. [Google Scholar] [CrossRef] [PubMed]
  50. Romani, A.; Bernini, R.; Noce, A.; Urciuoli, S.; Di Lauro, M.; Zaitseva, A.P.; Marrone, G.; Daniele, N. Di Potential Beneficial Effects of Extra Virgin Olive Oils Characterized by High Content in Minor Polar Compounds in Nephropathic Patients: A Pilot Study. Molecules 2020, 25, 4757. [Google Scholar] [CrossRef] [PubMed]
  51. Marrone, G.; Urciuoli, S.; Di Lauro, M.; Ruzzolini, J.; Ieri, F.; Vignolini, P.; Di Daniele, F.; Guerriero, C.; Nediani, C.; Di Daniele, N.; et al. Extra Virgin Olive Oil and Cardiovascular Protection in Chronic Kidney Disease. Nutrients 2022, 14, 4265. [Google Scholar] [CrossRef] [PubMed]
  52. Silva, S.; Bronze, M.R.; Figueira, M.E.; Siwy, J.; Mischak, H.; Combet, E.; Mullen, W. Impact of a 6-Wk Olive Oil Supplementation in Healthy Adults on Urinary Proteomic Biomarkers of Coronary Artery Disease, Chronic Kidney Disease, and Diabetes (Types 1 and 2): A Randomized, Parallel, Controlled, Double-Blind Study. Am. J. Clin. Nutr. 2015, 101, 44–54. [Google Scholar] [CrossRef] [PubMed]
  53. Jespersen, T.; Kruse, N.; Mehta, T.; Kuwabara, M.; Noureddine, L.; Jalal, D. Light Wine Consumption Is Associated with a Lower Odd for Cardiovascular Disease in Chronic Kidney Disease. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 1133–1139. [Google Scholar] [CrossRef]
  54. Monagas, M.; Bartolomé, B.; Gómez-Cordovés, C. Updated Knowledge about the Presence of Phenolic Compounds in Wine. Crit. Rev. Food Sci. Nutr. 2005, 45, 85–118. [Google Scholar] [CrossRef] [PubMed]
  55. Anvarifard, P.; Anbari, M.; Ostadrahimi, A.; Ardalan, M.; Ghoreishi, Z. Effects of Iranian Propolis on Renal Function, Prooxidant-Antioxidant Balance, Metabolic Status, and Quality of Life in Patients with Chronic Kidney Disease: A Study Protocol of an Ongoing Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Contemp. Clin. Trials Commun. 2023, 34, 101159. [Google Scholar] [CrossRef] [PubMed]
  56. Hammad, M.; Raftari, M.; Cesário, R.; Salma, R.; Godoy, P.; Emami, S.N.; Haghdoost, S. Roles of Oxidative Stress and Nrf2 Signaling in Pathogenic and Non-Pathogenic Cells: A Possible General Mechanism of Resistance to Therapy. Antioxidants 2023, 12, 1371. [Google Scholar] [CrossRef] [PubMed]
  57. Alvarenga, L.; Salarolli, R.; Cardozo, L.F.M.F.; Santos, R.S.; de Brito, J.S.; Kemp, J.A.; Reis, D.; de Paiva, B.R.; Stenvinkel, P.; Lindholm, B.; et al. Impact of Curcumin Supplementation on Expression of Inflammatory Transcription Factors in Hemodialysis Patients: A Pilot Randomized, Double-Blind, Controlled Study. Clin. Nutr. 2020, 39, 3594–3600. [Google Scholar] [CrossRef] [PubMed]
  58. Kanlaya, R.; Peerapen, P.; Nilnumkhum, A.; Plumworasawat, S.; Sueksakit, K.; Thongboonkerd, V. Epigallocatechin-3-Gallate Prevents TGF-Β1-Induced Epithelial-Mesenchymal Transition and Fibrotic Changes of Renal Cells via GSK-3β/β-Catenin/Snail1 and Nrf2 Pathways. J. Nutr. Biochem. 2020, 76, 108266. [Google Scholar] [CrossRef] [PubMed]
  59. Saldanha, J.F.; Leal, V.O.; Rizzetto, F.; Grimmer, G.H.; Ribeiro-Alves, M.; Daleprane, J.B.; Carraro-Eduardo, J.C.; Mafra, D. Effects of Resveratrol Supplementation in Nrf2 and NF-ΚB Expressions in Nondialyzed Chronic Kidney Disease Patients: A Randomized, Double-Blind, Placebo-Controlled, Crossover Clinical Trial. J. Ren. Nutr. 2016, 26, 401–406. [Google Scholar] [CrossRef] [PubMed]
  60. Chuang, S.T.; Kuo, Y.H.; Su, M.J. KS370G, a Caffeamide Derivative, Attenuates Unilateral Ureteral Obstruction-Induced Renal Fibrosis by the Reduction of Inflammation and Oxidative Stress in Mice. Eur. J. Pharmacol. 2015, 750, 1–7. [Google Scholar] [CrossRef] [PubMed]
  61. Sato, S.; Norikura, T.; Mukai, Y.; Yamaoka, S.; Mikame, K. Lignin-Derived Low-Molecular-Weight Oxidized Lignophenol Stimulates AMP-Activated Protein Kinase and Suppresses Renal Inflammation and Interstitial Fibrosis in High Fat Diet-Fed Mice. Chem. Biol. Interact. 2020, 318, 108977. [Google Scholar] [CrossRef] [PubMed]
  62. Ribeiro, M.; Fanton, S.; Paiva, B.R.; Baptista, B.G.; Alvarenga, L.; Ribeiro-Alves, M.; Cardozo, L.F.; Mafra, D. Dark Chocolate (70% Cocoa) Attenuates the Inflammatory Marker TNF-α in Patients on Hemodialysis. Clin. Nutr. ESPEN 2023, 53, 189–195. [Google Scholar] [CrossRef]
  63. Wollgast, J.; Anklam, E. Polyphenols in Chocolate: Is There a Contribution to Human Health? Food Res. Int. 2000, 33, 449–459. [Google Scholar] [CrossRef]
  64. Phrueksanan, W.; Yibchok-Anun, S.; Adisakwattana, S. Protection of Clitoria Ternatea Flower Petal Extract against Free Radicalinduced Hemolysis and Oxidative Damage in Canine Erythrocytes. Res. Vet. Sci. 2014, 97, 357–363. [Google Scholar] [CrossRef] [PubMed]
  65. López Prado, A.S.; Shen, Y.; Ardoin, R.; Osorio, L.F.; Cardona, J.; Xu, Z.; Prinyawiwatkul, W. Effects of Different Solvents on Total Phenolic and Total Anthocyanin Contents of Clitoria ternatea L. Petal and Their Anti-Cholesterol Oxidation Capabilities. Int. J. Food Sci. Technol. 2019, 54, 424–431. [Google Scholar] [CrossRef]
  66. Monteiro, E.B.; Soares, E.D.R.; Trindade, P.L.; de Bem, G.F.; Resende, A.D.C.; Passos, M.M.C.D.F.; Soulage, C.O.; Daleprane, J.B. Uraemic Toxin-Induced Inflammation and Oxidative Stress in Human Endothelial Cells: Protective Effect of Polyphenol-Rich Extract from Açaí. Exp. Physiol. 2020, 105, 542–551. [Google Scholar] [CrossRef] [PubMed]
  67. Pacheco-Palencia, L.A.; Mertens-Talcott, S.; Talcott, S.T. Chemical Composition, Antioxidant Properties, and Thermal Stability of a Phytochemical Enriched Oil from Açai (Euterpe Oleracea Mart.). J. Agric. Food Chem. 2008, 56, 4631–4636. [Google Scholar] [CrossRef]
  68. Zhou, S.; He, Y.; Zhang, W.; Xiong, Y.; Jiang, L.; Wang, J.; Cui, X.; Qu, Y.; Ge, F. Ophiocordyceps Lanpingensis Polysaccharides Alleviate Chronic Kidney Disease through MAPK/NF-ΚB Pathway. J. Ethnopharmacol. 2021, 276, 114189. [Google Scholar] [CrossRef]
  69. Zhang, Y.; Liu, Y.; Li, Y.; Zhang, L.; Li, C. Volatile components in mycelia and fruiting bodies of the artificial culture of Ophiocordyceps longissima. J. Anhui Agric. Univ. 2014, 41, 994–998. [Google Scholar]
  70. Vranješ, M.; Popović, B.M.; Štajner, D.; Ivetić, V.; Mandić, A.; Vranješ, D. Effects of Bearberry, Parsley and Corn Silk Extracts on Diuresis, Electrolytes Composition, Antioxidant Capacity and Histopathological Features in Mice Kidneys. J. Funct. Foods 2016, 21, 272–282. [Google Scholar] [CrossRef]
  71. Lopes, R.d.C.S.O.; de Lima, S.L.S.; da Silva, B.P.; Toledo, R.C.L.; Moreira, M.E.d.C.; Anunciação, P.C.; Walter, E.H.M.; Carvalho, C.W.P.; Queiroz, V.A.V.; Ribeiro, A.Q.; et al. Evaluation of the Health Benefits of Consumption of Extruded Tannin Sorghum with Unfermented Probiotic Milk in Individuals with Chronic Kidney Disease. Food Res. Int. 2018, 107, 629–638. [Google Scholar] [CrossRef]
  72. Conte, F.L.; Pereira, A.C.; Brites, G.; Ferreira, I.; Silva, A.C.; Sebastião, A.I.; Matos, P.; Pereira, C.; Batista, M.T.; Sforcin, J.M.; et al. Exploring the Antioxidant, Anti-Inflammatory and Antiallergic Potential of Brazilian Propolis in Monocytes. Phytomed. Plus 2022, 2, 100231. [Google Scholar] [CrossRef]
  73. Corredor, Z.; Rodríguez-Ribera, L.; Coll, E.; Montañés, R.; Diaz, J.M.; Ballarin, J.; Marcos, R.; Pastor, S. Unfermented Grape Juice Reduce Genomic Damage on Patients Undergoing Hemodialysis. Food Chem. Toxicol. 2016, 92, 1–7. [Google Scholar] [CrossRef]
  74. Aranda-Rivera, A.K.; Cruz-Gregorio, A.; Pedraza-Chaverri, J.; Scholze, A. Nrf2 Activation in Chronic Kidney Disease: Promises and Pitfalls. Antioxidants 2022, 11, 1112. [Google Scholar] [CrossRef] [PubMed]
  75. Jung, S.; Bae, H.; Song, W.S.; Jang, C. Dietary Fructose and Fructose-Induced Pathologies. Annu. Rev. Nutr. 2022, 42, 45–66. [Google Scholar] [CrossRef] [PubMed]
  76. Malik, V.S.; Popkin, B.M.; Bray, G.A.; Després, J.P.; Hu, F.B. Sugar-Sweetened Beverages, Obesity, Type 2 Diabetes Mellitus, and Cardiovascular Disease Risk. Circulation 2010, 121, 1356–1364. [Google Scholar] [CrossRef] [PubMed]
  77. Magazzù, A.; Marcuello, C. Investigation of Soft Matter Nanomechanics by Atomic Force Microscopy and Optical Tweezers: A Comprehensive Review. Nanomaterials 2023, 13, 963. [Google Scholar] [CrossRef] [PubMed]
  78. Chang, Z.; Zhang, L.; Hang, J.T.; Liu, W.; Xu, G.K. Viscoelastic Multiscale Mechanical Indexes for Assessing Liver Fibrosis and Treatment Outcomes. Nano Lett. 2023, 23, 9618–9625. [Google Scholar] [CrossRef] [PubMed]
  79. Abdel-Kawi, S.H.; Hassanin, K.M.A.; Hashem, K.S. The Effect of High Dietary Fructose on the Kidney of Adult Albino Rats and the Role of Curcumin Supplementation: A Biochemical and Histological Study. Beni-Suef Univ. J. Basic Appl. Sci. 2016, 5, 52–60. [Google Scholar] [CrossRef]
  80. Chung, A.P.Y.S.; Ton, S.H.; Gurtu, S.; Palanisamy, U.D. Ellagitannin Geraniin Supplementation Ameliorates Metabolic Risks in High-Fat Diet-Induced Obese Sprague Dawley Rats. J. Funct. Foods 2014, 9, 173–182. [Google Scholar] [CrossRef]
  81. Nour, O.A.; Ghoniem, H.A.; Nader, M.A.; Suddek, G.M. Impact of Protocatechuic Acid on High Fat Diet-Induced Metabolic Syndrome Sequelae in Rats. Eur. J. Pharmacol. 2021, 907, 174257. [Google Scholar] [CrossRef] [PubMed]
  82. de Sousa, A.R.; de Castro Moreira, M.E.; Grancieri, M.; Toledo, R.C.L.; de Oliveira Araújo, F.; Mantovani, H.C.; Queiroz, V.A.V.; Martino, H.S.D. Extruded Sorghum (Sorghum bicolor L.) Improves Gut Microbiota, Reduces Inflammation, and Oxidative Stress in Obese Rats Fed a High-Fat Diet. J. Funct. Foods 2019, 58, 282–291. [Google Scholar] [CrossRef]
  83. Noratto, G.D.; Murphy, K.; Chew, B.P. Quinoa Intake Reduces Plasma and Liver Cholesterol, Lessens Obesity-Associated Inflammation, and Helps to Prevent Hepatic Steatosis in Obese Db/Db Mouse. Food Chem. 2019, 287, 107–114. [Google Scholar] [CrossRef]
  84. Park, C.H.; Noh, J.S.; Yamabe, N.; Kang, K.S.; Tanaka, T.; Yokozawa, T. Beneficial Effect of 7-O-Galloyl-d-Sedoheptulose on Oxidative Stress and Hepatic and Renal Changes in Type 2 Diabetic Db/Db Mice. Eur. J. Pharmacol. 2010, 640, 233–242. [Google Scholar] [CrossRef] [PubMed]
  85. Saravanan, S.; Pari, L. Protective Effect of Thymol on High Fat Diet Induced Diabetic Nephropathy in C57BL/6J Mice. Chem. Biol. Interact. 2016, 245, 1–11. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, Y.S.; Huang, C.N.; Wang, C.J.; Lee, Y.J.; Chen, M.L.; Peng, C.H. Polyphenols of Hibiscus Sabdariffa Improved Diabetic Nephropathy via Regulating the Pathogenic Markers and Kidney Functions of Type 2 Diabetic Rats. J. Funct. Foods 2013, 5, 810–819. [Google Scholar] [CrossRef]
  87. Wu, C.C.; Hung, C.N.; Shin, Y.C.; Wang, C.J.; Huang, H.P. Myrciaria Cauliflora Extracts Attenuate Diabetic Nephropathy Involving the Ras Signaling Pathway in Streptozotocin/Nicotinamide Mice on a High Fat Diet. J. Food Drug Anal. 2016, 24, 136–146. [Google Scholar] [CrossRef] [PubMed]
  88. Mahajan, R.; Prasad, S.; Gaikwad, S.; Itankar, P. Antioxidant Phenolic Compounds from Seeds of Hordeum Vulgare Linn. Ameliorates Diabetic Nephropathy in Streptozotocin-Induced Diabetic Rats. J. Tradit. Chin. Med. Sci. 2023, 10, 353–361. [Google Scholar] [CrossRef]
  89. Da-Costa-Rocha, I.; Bonnlaender, B.; Sievers, H.; Pischel, I.; Heinrich, M. Hibiscus sabdariffa L.—A Phytochemical and Pharmacological Review. Food Chem. 2014, 165, 424–443. [Google Scholar] [CrossRef] [PubMed]
  90. Gansevoort, R.T.; Correa-Rotter, R.; Hemmelgarn, B.R.; Jafar, T.H.; Heerspink, H.J.L.; Mann, J.F.; Matsushita, K.; Wen, C.P. Chronic Kidney Disease and Cardiovascular Risk: Epidemiology, Mechanisms, and Prevention. Lancet 2013, 382, 339–352. [Google Scholar] [CrossRef] [PubMed]
  91. Poznyak, A.V.; Sadykhov, N.K.; Kartuesov, A.G.; Borisov, E.E.; Sukhorukov, V.N.; Orekhov, A.N. Atherosclerosis Specific Features in Chronic Kidney Disease (CKD). Biomedicines 2022, 10, 2094. [Google Scholar] [CrossRef] [PubMed]
  92. Emamat, H.; Zahedmehr, A.; Asadian, S.; Nasrollahzadeh, J. The Effect of Purple-Black Barberry (Berberis Integerrima) on Blood Pressure in Subjects with Cardiovascular Risk Factors: A Randomized Controlled Trial. J. Ethnopharmacol. 2022, 289, 115097. [Google Scholar] [CrossRef]
  93. Gomes, A.; Godinho-Pereira, J.; Oudot, C.; Sequeira, C.O.; Macià, A.; Carvalho, F.; Motilva, M.J.; Pereira, S.A.; Matzapetakis, M.; Brenner, C.; et al. Berry Fruits Modulate Kidney Dysfunction and Urine Metabolome in Dahl Salt-Sensitive Rats. Free Radic. Biol. Med. 2020, 154, 119–131. [Google Scholar] [CrossRef]
  94. Memije-Lazaro, I.N.; Blas-Valdivia, V.; Franco-Colín, M.; Cano-Europa, E. Arthrospira Maxima (Spirulina) and C-Phycocyanin Prevent the Progression of Chronic Kidney Disease and Its Cardiovascular Complications. J. Funct. Foods 2018, 43, 37–43. [Google Scholar] [CrossRef]
  95. Sonfack, C.S.; Nguelefack-Mbuyo, E.P.; Kojom, J.J.; Lappa, E.L.; Peyembouo, F.P.; Fofié, C.K.; Nolé, T.; Nguelefack, T.B.; Dongmo, A.B. The Aqueous Extract from the Stem Bark of Garcinia Lucida Vesque (Clusiaceae) Exhibits Cardioprotective and Nephroprotective Effects in Adenine-Induced Chronic Kidney Disease in Rats. Evid.-Based Complement. Altern. Med. 2021, 2021, 5581041. [Google Scholar] [CrossRef] [PubMed]
  96. da Silva, M.F.; Casazza, A.A.; Ferrari, P.F.; Aliakbarian, B.; Converti, A.; Bezerra, R.P.; Porto, A.L.F.; Perego, P. Recovery of Phenolic Compounds of Food Concern from Arthrospira Platensis by Green Extraction Techniques. Algal Res. 2017, 25, 391–401. [Google Scholar] [CrossRef]
  97. Fotie, J.; Bohle, D.S.; Olivier, M.; Gomez, M.A.; Nzimiro, S. Trypanocidal and Antileishmanial Dihydrochelerythrine Derivatives from Garcinia Lucida. J. Nat. Prod. 2007, 70, 1650–1653. [Google Scholar] [CrossRef] [PubMed]
  98. Campidelli, M.L.L.; Carneiro, J.D.S.; Souza, E.C.; Magalhães, M.L.; Nunes, E.E.C.; Faria, P.B.; Franco, M.; Boas, E.V.B.V. Effects of the Drying Process on the Fatty Acid Content, Phenolic Profile, Tocopherols and Antioxidant Activity of Baru Almonds (Dipteryx Alata Vog.). Grasas Aceites 2020, 71, e343. [Google Scholar] [CrossRef]
  99. Kpemissi, M.; Veerapur, V.P.; Suhas, D.S.; Puneeth, T.A.; Nandeesh, R.; Vijayakumar, S.; Eklu-Gadegbeku, K. Combretum Micranthum G. Don Protects Hypertension Induced by L-NAME by Cardiovascular and Renal Remodelling through Reversing Inflammation and Oxidative Stress. J. Funct. Foods 2022, 94, 105132. [Google Scholar] [CrossRef]
  100. Mamun, F.; Rahman, M.M.; Zamila, M.; Subhan, N.; Hossain, H.; Raquibul Hasan, S.M.; Alam, M.A.; Haque, M.A. Polyphenolic Compounds of Litchi Leaf Augment Kidney and Heart Functions in 2K1C Rats. J. Funct. Foods 2020, 64, 103662. [Google Scholar] [CrossRef]
  101. Schincaglia, R.M.; Cuppari, L.; Neri, H.F.S.; Cintra, D.E.; Sant’Ana, M.R.; Mota, J.F. Effects of Baru Almond Oil (Dipteryx Alata Vog.) Supplementation on Body Composition, Inflammation, Oxidative Stress, Lipid Profile, and Plasma Fatty Acids of Hemodialysis Patients: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Complement. Ther. Med. 2020, 52, 102479. [Google Scholar] [CrossRef] [PubMed]
  102. Vertolli, U.; Davis, P.A.; Maso, L.D.; Maiolino, G.; Naso, A.; Plebani, M.; Calò, L.A. Daily Green Tea Extract Supplementation Reduces Prothrombotic and Inflammatory States in Dialysis Patients. J. Funct. Foods 2013, 5, 1366–1371. [Google Scholar] [CrossRef]
  103. Calo, L.A.; Vertolli, U.; Davis, P.A.; Maso, L.D.; Pagnin, E.; Ravarotto, V.; Maiolino, G.; Lupia, M.; Seccia, T.M.; Rossi, G.P. Molecular Biology Based Assessment of Green Tea Effects on Oxidative Stress and Cardiac Remodelling in Dialysis Patients. Clin. Nutr. 2014, 33, 437–442. [Google Scholar] [CrossRef]
  104. Lew, Q.L.J.; Jafar, T.H.; Jin, A.; Yuan, J.M.; Koh, W.P. Consumption of Coffee but Not of Other Caffeine-Containing Beverages Reduces the Risk of End-Stage Renal Disease in the Singapore Chinese Health Study. J. Nutr. 2018, 148, 1315–1322. [Google Scholar] [CrossRef] [PubMed]
  105. Pazhayattil, G.S.; Shirali, A.C. Drug-Induced Impairment of Renal Function. Int. J. Nephrol. Renovasc. Dis. 2014, 7, 457–468. [Google Scholar] [CrossRef] [PubMed]
  106. Dobrek, L. A Synopsis of Current Theories on Drug-Induced Nephrotoxicity. Life 2023, 13, 325. [Google Scholar] [CrossRef] [PubMed]
  107. Ungur, R.; Buzatu, R.; Lacatus, R.; Purdoiu, R.C.; Petrut, G.; Codea, R.; Sarpataky, O.; Biris, A.; Popovici, C.; Mircean, M.; et al. Evaluation of the Nephroprotective Effect of Sambucus Nigra Total Extract in a Rat Experimental Model of Gentamicine Nephrotoxicity. Rev. Chim. 2019, 70, 1971–1974. [Google Scholar] [CrossRef]
  108. El-Ashmawy, N.E.; Khedr, N.F.; El-Bahrawy, H.A.; Helal, S.A. Upregulation of PPAR-γ Mediates the Renoprotective Effect of Omega-3 PUFA and Ferulic Acid in Gentamicin-Intoxicated Rats. Biomed. Pharmacother. 2018, 99, 504–510. [Google Scholar] [CrossRef] [PubMed]
  109. Rashid, S.; Ali, N.; Nafees, S.; Ahmad, S.T.; Hasan, S.K.; Sultana, S. Abrogation of 5-Flourouracil Induced Renal Toxicity by Bee Propolis via Targeting Oxidative Stress and Inflammation in Wistar Rats. J. Pharm. Res. 2013, 7, 189–194. [Google Scholar] [CrossRef]
  110. Búfalo, M.C.; Ferreira, I.; Costa, G.; Francisco, V.; Liberal, J.; Cruz, M.T.; Lopes, M.C.; Batista, M.T.; Sforcin, J.M. Propolis and Its Constituent Caffeic Acid Suppress LPS-Stimulated pro-Inflammatory Response by Blocking NF-ΚB and MAPK Activation in Macrophages. J. Ethnopharmacol. 2013, 149, 84–92. [Google Scholar] [CrossRef] [PubMed]
  111. El-Sayed, S.M.; El-Naggar, M.E.; Hussein, J.; Medhat, D.; El-Banna, M. Effect of Ficus carica L. Leaves Extract Loaded Gold Nanoparticles against Cisplatin-Induced Acute Kidney Injury. Colloids Surfaces B Biointerfaces 2019, 184, 110465. [Google Scholar] [CrossRef] [PubMed]
  112. Veberic, R.; Colaric, M.; Stampar, F. Phenolic Acids and Flavonoids of Fig Fruit (Ficus carica L.) in the Northern Mediterranean Region. Food Chem. 2008, 106, 153–157. [Google Scholar] [CrossRef]
  113. Al-Okbi, S.Y.; Mohamed, D.A.; Hamed, T.E.; Esmail, R.S.H.; Donya, S.M. Prevention of Renal Dysfunction by Nutraceuticals Prepared from Oil Rich Plant Foods. Asian Pac. J. Trop. Biomed. 2014, 4, 618–626. [Google Scholar] [CrossRef]
  114. Corbin, C.; Fidel, T.; Leclerc, E.A.; Barakzoy, E.; Sagot, N.; Falguiéres, A.; Renouard, S.; Blondeau, J.P.; Ferroud, C.; Doussot, J.; et al. Development and Validation of an Efficient Ultrasound Assisted Extraction of Phenolic Compounds from Flax (Linum usitatissimum L.) Seeds. Ultrason. Sonochem. 2015, 26, 176–185. [Google Scholar] [CrossRef] [PubMed]
  115. Ramírez-Rodríguez, Y.; Ramírez, V.; Robledo-Márquez, K.; García-Rojas, N.; Rojas-Morales, P.; Arango, N.; Pedraza-Chaverri, J.; Medina-Campos, O.N.; Pérez-Rojas, J.M.; Flores-Ramírez, R.; et al. Stenocereus Huastecorum-Fruit Juice Concentrate Protects against Cisplatin-Induced Nephrotoxicity by Nitric Oxide Pathway Activity and Antioxidant and Antiapoptotic Effects. Food Res. Int. 2022, 160, 111337. [Google Scholar] [CrossRef] [PubMed]
  116. Widowati, W.; Prahastuti, S.; Hidayat, M.; Hasianna, S.T.; Wahyudianingsih, R.; Eltania, T.F.; Azizah, A.M.; Aviani, J.K.; Subangkit, M.; Handayani, R.A.S.; et al. Detam 1 Black Soybean against Cisplatin-Induced Acute Ren Failure on Rat Model via Antioxidant, Antiinflammatory and Antiapoptosis Potential. J. Tradit. Complement. Med. 2022, 12, 426–435. [Google Scholar] [CrossRef]
  117. Amarasiri, S.S.; Attanayake, A.P.; Arawwawala, L.D.A.M.; Mudduwa, L.K.B.; Jayatilaka, K.A.P.W. Barleria prionitis L. Extracts Ameliorate Doxorubicin-Induced Acute Kidney Injury via Modulation of Oxidative Stress, Inflammation, and Apoptosis. J. Tradit. Complement. Med. 2023, 13, 500–510. [Google Scholar] [CrossRef]
  118. Amarasiri, S.S.; Attanayake, A.P.; Mudduwa, L.K.B.; Jayatilaka, K.A.P.W. Nephroprotective Mechanisms of Ambrette (Abelmoschus Moschatus Medik.) Leaf Extracts in Adriamycin Mediated Acute Kidney Injury Model of Wistar Rats. J. Ethnopharmacol. 2022, 292, 115221. [Google Scholar] [CrossRef] [PubMed]
  119. Amarasiri, S.S.; Attanayake, A.P.; Arawwawala, L.D.A.M.; Jayatilaka, K.A.P.W.; Mudduwa, L.K.B. Protective Effects of Three Selected Standardized Medicinal Plant Extracts Used in Sri Lankan Traditional Medicine in Adriamycin Induced Nephrotoxic Wistar Rats. J. Ethnopharmacol. 2020, 259, 112933. [Google Scholar] [CrossRef]
  120. Rahman, M.M.; Zaman, S.; Mamun, F.; Gias, Z.T.; Alam, M.N.; Ulla, A.; Hossain, M.H.; Reza, H.M.; Alam, M.A. Phenolic Content Analysis in Psidium Guajava Leaves Powder by HPLC-DAD System and in Vivo Renoprotective and Antioxidant Activities in Fludrocortisone Acetate-Induced Rats. J. Food Biochem. 2018, 42, 1–14. [Google Scholar] [CrossRef]
  121. Mehmood, A.; Zhao, L.; Ishaq, M.; Zad, O.D.; Zhao, L.; Wang, C.; Usman, M.; Lian, Y.; Xu, M. Renoprotective Effect of Stevia Residue Extract on Adenine-Induced Chronic Kidney Disease in Mice. J. Funct. Foods 2020, 72, 103983. [Google Scholar] [CrossRef]
  122. Covarrubias-Cárdenas, A.G.; Martínez-Castillo, J.I.; Medina-Torres, N.; Ayora-Talavera, T.; Espinosa-Andrews, H.; García-Cruz, N.U.; Pacheco, N. Antioxidant Capacity and Uplc-Pda Esi-Ms Phenolic Profile of Stevia Rebaudiana Dry Powder Extracts Obtained by Ultrasound Assisted Extraction. Agronomy 2018, 8, 170. [Google Scholar] [CrossRef]
  123. Adekunle, I.A.; Imafidon, C.E.; Oladele, A.A.; Ayoka, A.O. Ginger Polyphenols Attenuate Cyclosporine-Induced Disturbances in Kidney Function: Potential Application in Adjuvant Transplant Therapy. Pathophysiology 2018, 25, 101–115. [Google Scholar] [CrossRef]
  124. Selim, S.; Akter, N.; Nayan, S.I.; Chowdhury, F.I.; Saffoon, N.; Khan, F.; Ahmed, K.S.; Ahmed, M.I.; Hossain, M.M.; Alam, M.A. Flacourtia Indica Fruit Extract Modulated Antioxidant Gene Expression, Prevented Oxidative Stress and Ameliorated Kidney Dysfunction in Isoprenaline Administered Rats. Biochem. Biophys. Rep. 2021, 26, 101012. [Google Scholar] [CrossRef] [PubMed]
  125. Bojan, M.S.; Rajappa, R.; Vijayakumar, D.R.K.; Gopalan, J. Protective Effect of Raphanus Sativus on D-Galactosamine Induced Nephrotoxicity in Rats. Nutr. Clin. Metab. 2016, 30, 22–28. [Google Scholar] [CrossRef]
  126. Sgherri, C.; Cosi, E.; Navari-Izzo, F. Phenols and Antioxidative Status of Raphanus Sativus Grown in Copper Excess. Physiol. Plant. 2003, 118, 21–28. [Google Scholar] [CrossRef] [PubMed]
  127. Abdel-Rahman Mohamed, A.; El-Kholy, S.S.; Dahran, N.; El Bohy, K.M.; Moustafa, G.G.; Saber, T.M.; Metwally, M.M.M.; Gaber, R.A.; Alqahtani, L.S.; Mostafa-Hedeab, G.; et al. Scrutinizing Pathways of Nicotine Effect on Renal Alpha-7 Nicotinic Acetylcholine Receptor and Mitogen-Activated Protein Kinase (MAPK) Expression in Ehrlich Ascites Carcinoma-Bearing Mice: Role of Chlorella Vulgaris. Gene 2022, 837, 146697. [Google Scholar] [CrossRef] [PubMed]
  128. Shanab, O.; El-Rayes, S.M.; Khalil, W.F.; Ahmed, N.; Abdelkader, A.; Aborayah, N.H.; Atwa, A.M.; Mohammed, F.I.; Nasr, H.E.; Ibrahim, S.F.; et al. Nephroprotective Effects of Acacia Senegal against Aflatoxicosis via Targeting Inflammatory and Apoptotic Signaling Pathways. Ecotoxicol. Environ. Saf. 2023, 262, 115194. [Google Scholar] [CrossRef] [PubMed]
  129. Alomar, M.Y. Physiological and Histopathological Study on the Influence of Ocimum Basilicum Leaves Extract on Thioacetamide-Induced Nephrotoxicity in Male Rats. Saudi J. Biol. Sci. 2020, 27, 1843–1849. [Google Scholar] [CrossRef] [PubMed]
  130. Javanmardi, J.; Khalighi, A.; Kashi, A.; Bais, H.P.; Vivanco, J.M. Chemical Characterization of Basil (Ocimum basilicum L.) Found in Local Accessions and Used in Traditional Medicines in Iran. J. Agric. Food Chem. 2002, 50, 5878–5883. [Google Scholar] [CrossRef] [PubMed]
  131. Fathy, M.; Abdel-latif, R.; Abdelgwad, Y.M.; Othman, O.A.; Abdel-Razik, A.R.H.; Dandekar, T.; Othman, E.M. Nephroprotective Potential of Eugenol in a Rat Experimental Model of Chronic Kidney Injury; Targeting NOX, TGF-β, and Akt Signaling. Life Sci. 2022, 308, 120957. [Google Scholar] [CrossRef]
  132. Al-Asmari, K.M.; Altayb, H.N.; Al-Attar, A.M.; Qahl, S.H.; Al-Thobaiti, S.A.; Abu Zeid, I.M. Arabica Coffee and Olive Oils Mitigate Malathion-Induced Nephrotoxicity in Rat: In Silico, Immunohistochemical and Biochemical Evaluation. Saudi J. Biol. Sci. 2022, 29, 103307. [Google Scholar] [CrossRef]
  133. Emmanuel, O.; Okezie, U.M.; Iweala, E.J.; Ugbogu, E.A. Pretreatment of Red Palm Oil Extracted from Palm Fruit (Elaeis Guineensis) Attenuates Carbon Tetrachloride Induced Toxicity in Wistar Rats. Phytomed. Plus 2021, 1, 100079. [Google Scholar] [CrossRef]
  134. Tsouko, E.; Alexandri, M.; Fernandes, K.V.; Freire, D.M.G.; Mallouchos, A.; Koutinas, A.A. Extraction of Phenolic Compounds from Palm Oil Processing Residues and Their Application as Antioxidants. Food Technol. Biotechnol. 2019, 57, 29–38. [Google Scholar] [CrossRef] [PubMed]
  135. Aqeel, T.; Gurumallu, S.C.; Bhaskar, A.; Hashimi, S.M.; Javaraiah, R. Secoisolariciresinol Diglucoside Protects against Cadmium-Induced Oxidative Stress-Mediated Renal Toxicity in Rats. J. Trace Elem. Med. Biol. 2020, 61, 126552. [Google Scholar] [CrossRef] [PubMed]
  136. Yun, S.; Chu, D.; He, X.; Zhang, W.; Feng, C. Protective Effects of Grape Seed Proanthocyanidins against Iron Overload-Induced Renal Oxidative Damage in Rats. J. Trace Elem. Med. Biol. 2020, 57, 126407. [Google Scholar] [CrossRef] [PubMed]
  137. Fernando, T.D.; Jayawardena, B.M.; Mathota Arachchige, Y.L.N. Variation of Different Metabolites and Heavy Metals in Oryza sativa L., Related to Chronic Kidney Disease of Unknown Etiology in Sri Lanka. Chemosphere 2020, 247, 125836. [Google Scholar] [CrossRef] [PubMed]
  138. Tian, Y.; Lin, L.; Zhao, M.; Peng, A.; Zhao, K. Xanthine Oxidase Inhibitory Activity and Antihyperuricemic Effect of Moringa Oleifera Lam. Leaf Hydrolysate Rich in Phenolics and Peptides. J. Ethnopharmacol. 2021, 270, 113808. [Google Scholar] [CrossRef]
  139. Kang, L.; Miao, J.X.; Cao, L.H.; Miao, Y.Y.; Miao, M.S.; Liu, H.J.; Xiang, L.L.; Song, Y.G. Total Glucosides of Herbaceous Peony (Paeonia lactiflora Pall.) Flower Attenuate Adenine- and Ethambutol-Induced Hyperuricaemia in Rats. J. Ethnopharmacol. 2020, 261, 113054. [Google Scholar] [CrossRef] [PubMed]
  140. Chonghui, L.; Hui, D.; Wang, L.; Qingyan, S.; Zheng, Y.; Yanjun, X.; Zhang, J.; Zhang, J.; Yang, R.; Yuxuan, G. Flavonoid Composition and Antioxidant Activity of Tree Peony (Paeonia Section Moutan) Yellow Flowers. J. Agric. Food Chem. 2009, 57, 8496–8503. [Google Scholar] [CrossRef] [PubMed]
  141. Novaes, A.D.S.; Da Silva Mota, J.; Barison, A.; Veber, C.L.; Negrão, F.J.; Kassuya, C.A.L.; De Barros, M.E. Diuretic and Antilithiasic Activities of Ethanolic Extract from Piper Amalago (Piperaceae). Phytomedicine 2014, 21, 523–528. [Google Scholar] [CrossRef] [PubMed]
  142. Bawari, S.; Sah, A.N.; Tewari, D. Anticalcifying Effect of Daucus Carota in Experimental Urolithiasis in Wistar Rats. J. Ayurveda Integr. Med. 2020, 11, 308–315. [Google Scholar] [CrossRef]
  143. Bawari, S.; Sah, A.N.; Gupta, P.; Zengin, G.; Tewari, D. Himalayan Citrus Jambhiri Juice Reduced Renal Crystallization in Nephrolithiasis by Possible Inhibition of Glycolate Oxidase and Matrix Metalloproteinases. J. Ethnopharmacol. 2023, 306, 116157. [Google Scholar] [CrossRef]
  144. Moreno, K.G.T.; Gasparotto Junior, A.; dos Santos, A.C.; Palozi, R.A.C.; Guarnier, L.P.; Marques, A.A.M.; Romão, P.V.M.; Lorençone, B.R.; Cassemiro, N.S.; Silva, D.B.; et al. Nephroprotective and Antilithiatic Activities of Costus Spicatus (Jacq.) Sw.: Ethnopharmacological Investigation of a Species from the Dourados Region, Mato Grosso Do Sul State, Brazil. J. Ethnopharmacol. 2021, 266, 113409. [Google Scholar] [CrossRef] [PubMed]
  145. Younis, W.; Alamgeer; Schini-Kerth, V.B.; Brentan da Silva, D.; Junior, A.G.; Bukhari, I.A.; Assiri, A.M. Role of the NO/CGMP Pathway and Renin-Angiotensin System in the Hypotensive and Diuretic Effects of Aqueous Soluble Fraction from Crataegus Songarica K. Koch. J. Ethnopharmacol. 2020, 249, 112400. [Google Scholar] [CrossRef] [PubMed]
  146. Cao, Y.L.; Lin, J.H.; Hammes, H.P.; Zhang, C. Flavonoids in Treatment of Chronic Kidney Disease. Molecules 2022, 27, 2365. [Google Scholar] [CrossRef] [PubMed]
  147. Spiegel, M.; Kapusta, K.; Kołodziejczyk, W.; Saloni, J.; Zbikowska, B.; Hill, G.A.; Sroka, Z. Antioxidant Activity of Selected Phenolic Acids–Ferric Reducing Antioxidant Power Assay and QSAR Analysis of the Structural Features. Molecules 2020, 25, 3088. [Google Scholar] [CrossRef] [PubMed]
  148. Santos, C.M.M.; Silva, A.M.S.; Filipe, P.; Santus, R.; Patterson, L.K.; Mazière, J.C.; Cavaleiro, J.A.S.; Morlière, P. Structure-Activity Relationships in Hydroxy-2,3-Diarylxanthone Antioxidants. Fast Kinetics Spectroscopy as a Tool to Evaluate the Potential for Antioxidant Activity in Biological Systems. Org. Biomol. Chem. 2011, 9, 3965–3974. [Google Scholar] [CrossRef] [PubMed]
  149. Pavlatou, M.G.; Papastamataki, M.; Apostolakou, F.; Papassotiriou, I.; Tentolouris, N. FORT and FORD: Two Simple and Rapid Assays in the Evaluation of Oxidative Stress in Patients with Type 2 Diabetes Mellitus. Metabolism 2009, 58, 1657–1662. [Google Scholar] [CrossRef] [PubMed]
  150. Rubio, C.P.; Hernández-Ruiz, J.; Martinez-Subiela, S.; Tvarijonaviciute, A.; Ceron, J.J. Spectrophotometric Assays for Total Antioxidant Capacity (TAC) in Dog Serum: An Update. BMC Vet. Res. 2016, 12, 166. [Google Scholar] [CrossRef] [PubMed]
  151. Nguyen, D.D.; Luo, L.J.; Lue, S.J.; Lai, J.Y. The Role of Aromatic Ring Number in Phenolic Compound-Conjugated Chitosan Injectables for Sustained Therapeutic Antiglaucoma Efficacy. Carbohydr. Polym. 2020, 231, 115770. [Google Scholar] [CrossRef] [PubMed]
  152. Bukowska, B.; Michałowicz, J.; Krokosz, A.; Sicińska, P. Comparison of the Effect of Phenol and Its Derivatives on Protein and Free Radical Formation in Human Erythrocytes (in Vitro). Blood Cells Mol. Dis. 2007, 39, 238–244. [Google Scholar] [CrossRef]
  153. Beckman, J.S.; Ischiropoulos, H.; Zhu, L.; van der Woerd, M.; Smith, C.; Chen, J.; Harrison, J.; Martin, J.C.; Tsai, M. Kinetics of Superoxide Dismutase- and Iron-Catalyzed Nitration of Phenolics by Peroxynitrite. Arch. Biochem. Biophys. 1992, 298, 438–445. [Google Scholar] [CrossRef]
  154. Chen, Y.; Xiao, H.; Zheng, J.; Liang, G. Structure-Thermodynamics-Antioxidant Activity Relationships of Selected Natural Phenolic Acids and Derivatives: An Experimental and Theoretical Evaluation. PLoS ONE 2015, 10, e0121276. [Google Scholar] [CrossRef] [PubMed]
  155. Ávila-Román, J.; Soliz-Rueda, J.R.; Bravo, F.I.; Aragonès, G.; Suárez, M.; Arola-Arnal, A.; Mulero, M.; Salvadó, M.J.; Arola, L.; Torres-Fuentes, C.; et al. Phenolic Compounds and Biological Rhythms: Who Takes the Lead? Trends Food Sci. Technol. 2021, 113, 77–85. [Google Scholar] [CrossRef]
  156. Hajam, Y.A.; Rai, S.; Kumar, R.; Bashir, M.; Malik, J.A. Phenolic Compounds from Medicinal Herbs: Their Role in Animal Health and Diseases—A New Approach for Sustainable Welfare and Development. Plant Phenolics Sustain. Agric. 2020, 1, 221–239. [Google Scholar] [CrossRef]
  157. Kumar, N.; Goel, N. Phenolic Acids: Natural Versatile Molecules with Promising Therapeutic Applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chronic kidney disease-related conditions.
Figure 1. Chronic kidney disease-related conditions.
Molecules 29 02576 g001
Figure 2. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only.
Figure 2. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only.
Molecules 29 02576 g002
Figure 3. Oxidative stress: Nrf2 signaling pathway (adapted from Hammad et al. [56]).
Figure 3. Oxidative stress: Nrf2 signaling pathway (adapted from Hammad et al. [56]).
Molecules 29 02576 g003
Table 1. Phenolic compounds and their reported bioactivity in the reviewed studies.
Table 1. Phenolic compounds and their reported bioactivity in the reviewed studies.
Compound Reported Bioactivity
Hydroxytyrosol [50]↑ e-GFR after 9 weeks
↓ oxidative stress
↓ uric acid levels
[50]
Oleacin (10-hydroxy-oleocanthal)
Hydroxytyrosol
Tyrosol
Elenolic acid (and derivatives)
[51]↓ serum levels of creatinine, azotemia, albuminuria, uric acid
↓ FORT level, ESR, and CRP, TNF-α, and IL-6 serum levels
[51]
Hydroxytyrosol (and derivatives)[52]Improvement in the urinary proteomic biomarker of disease over a relatively short period in a healthy population[52]
Hydroxybenzoic and hydroxycinnamic acids
Stilbenes
[54]↓ hypertension, HDLs[53]
Curcumin[57]↓ NF-kB mRNA expression and hsCRP plasma levels[57]
Epigallocatechin-3-gallate[58]Prevention of EMT in renal tubular cells induced by the crosstalk between TGF-β1 and β-catenin signaling[58]
Resveratrol[59]There was no effect of resveratrol supplementation on Nfr2 and NF-kB expression at the dosage tested[59]
Caffeamide (derivative)[60]↓ oxidative stress
↓ AngII and TGF-β1 production and Smad3 phosphorylation
[60]
Lignophenol[61]↓ plasma BUN levels
↓ TNF-α, Ccl2, and TGF-β
[61]
Flavan-3-ols[63]↓ plasma levels of TNF-a[62]
Kaempferol
Caffeoylmalic acid
[65]Prevented a decrease in GSH concentration in erythrocytes
Inhibited the formation of the echinocytic form
[64]
Vanillic acid, syringic acid, p-hydroxybenzoic acid, protocatechuic acid, ferulic acid, and (+)-catechin [67]Prevented higher expression of MCP-1 and ICAM-1
↓ TNF-α concentration in cells
↓ protein carbonylation
[66]
1-octene-3-alcohl and butylated hydroxytoluene [69]↓ BUN and Scr
↓ content of ROS in renal tissues
↓ oxidative stress
↑ SOD and GSH-PX
[68]
Gallic acid, catechin and derivatives, and rutin and derivatives[70]↓ blood urea nitrogen and serum creatinine[70]
Luteolinidin and 5- methoxyluteolinidine [71]↓ oxidative stress
↑ serum SOD and TAC
↓ serum MDA decreased
[71]
Hydroxycinnamic acids and flavonoids and caffeic acid derivatives[110]↑ SOD 1 activity
↓ IL-1β, TNF-α, and IL-6 production
[72]
Hydroxybenzoic and hydroxycinnamic acids, stilbenes [54]↓ LDLs and cholesterol
↓ oxidative DNA damage
[73]
Curcumin[79]↓ serum urea and creatinine
↓ HO1 and INOS mRNA expression
↑ GSH concentration, GR, CAT, and SOD activities
↓ LPO and DNA fragmentation
[79]
Ellagitannin geraniin[80]↓ relative weights of pancreas, liver, heart, and aorta
↓ plasma glucose
↓ TG, non-HDLs and total cholesterols, ALT, AST, CK, and Cr
[80]
Protocatechuic acid[81]↓ BMI
↓ SOD activity in liver and muscle homogenates
↑ GSH activity in muscle
↓ MCP-1, IL-1b, CRP
[81]
3,4-dihydroxybenzoic acid [82]↓ TNF-α expression by 0.12 times[82]
Ferulic and benzoic acid derivatives, quercetin derivatives, flavonone glycosides[83]↓ cholesterol, LDLs, and plasma TGCs[83]
7-O-Galloyl-d-Sedoheptulose[84]↓ serum levels of triglycerides, total cholesterol, LDL/VLDL-cholesterol, NEFAs, and TBA-reactive substances
↓ serum levels of ALT, AST, creatinine, and urea nitrogen
[84]
Thymol[85]↓ urinary glucose, urinary urea, and urinary protein
↓ TBARS and LOOH
↑ SOD, catalase, GPx, GST, and GR
↓ SREBP-1c, TGF-b, and VEGF proteins
[85]
Protocatechuic acid[89]↓ insulin
Improved AGE expression and histological changes in both glomerular hypertrophy and interstitial crushing
[86]
Protocatechuic acid, gallic acid, catechin, gallocatechin, and rutin[87]↓ triacylglycerol and total cholesterol levels
↓ BUN, blood creatinine, and blood pressure
↓ ratio of urine albumin/urine creatinine
Ameliorated mesangial fibrosis in part via the Ras/PI3K/Akt signaling pathway.
[87]
Epicatechin[88]↓ BUN, total cholesterol, and triglycerides
↓ MDA
↑ SOD, GSH, and catalyze
Regeneration of tubular epithelium, inhibition of necrosis and hemorrhages, and recovery of atrophic glomeruli
[88]
Gallic acid, catechin and derivatives, and rutin and derivatives [70]↓ systolic and mean arterial BP
↑ plasma and urinary nitrite and nitrate
[92]
Gallic acid, catechin and derivatives, and rutin and derivatives[70]Recovery in the kidney morphology[93]
Catechin[96]↑ GSH and the GSH2/GSSG ratio[94]
Stigmasterol, betulinic acid[97]↓ lipid peroxidation
Antihemolytic effect
↓ blood pressure
↓ kidney hypertrophy
↑ CAT and SOD in the kidney
[95]
Hyperoside, quercitrin, caftaric acid, gentisic acid, caffeic acid and chlorogenic acid[99]↓ blood pressure
↓ blood levels of uric acid, urea, creatinine, and urine levels of NAG
↓ TNF-α
[99]
Gallic acid, catechin, epicatechin, rutin, quercetin, and kaempferol [100]↓ plasma concentrations of ALT, AST, and ALP
↑ SOD
↓ MDA, NO, and APOP
↑ catalase and SOD
[100]
Caffeic acid, chlorogenic acid, anthocyanins, p-coumaric acid, ferulic acid, o-coumaric acid, quercetin, gallic acid, rutin, catechin[98]↓ CRP
The activity of SOD and catalase enzymes, as well as the concentration of MDA, did not differ from the placebo group
[101]
Epigallocatechin, catechin, and epicatechin gallate[102]↓ fibrinogen levels, protein expression of p22phox, and hsCPR[102]
Epigallocatechin, catechin, and epicatechin gallate [102]↓ pERK1/2 phosphorylation
↓ oxLDL plasma levels
[103]
Catechin, epicatechin, quercetin, caffeic acid, and ferulic acid[107]↓ urinary NAG activity
↓ renal histopathological lesion
↓ urea and serum creatinine
[107]
Ferulic acid ↓ serum BUN and creatinine levels
↓ urinary albumin concentration and urinary NAG activity
↑ CAT activity
↑ RvE1 concentration
↑ PPAR-γ gene expression
[108]
Ferulic acid[110]Prevented disruption of the normal renal architecture
↓ creatinine, BUN, LDH, TNFa, and KIM-1 levels
[109]
Gallic acid, chlorogenic acid, syringic acid, (+)-catechin, (β)-epicatechin, and rutin[112]↓ MDA, Hyp, and Hcy[111]
P-couramic, caffeic, and ferulic acids[114]↓ serum MDA, lipid peroxidation
↑ plasma catalase and total antioxidant capacity
↑ Cr
↓ chromosomal aberrations in bone marrow cells and sperm shape abnormalities
[113]
Betacyanins[115]↓ plasma creatinine, BUN, and NGAL
↓ tubular damage
↑ MDA levels
↓ Nrf2 levels
↑ NO2/NO3 levels
[115]
Isoflavones (daidzein, daidzin, genistin, biochanin A, and glycitein)[116]↓ cytoplasmic IFN- γ expression
↓ Casp-3 expression
↓ IL-6, IL-1b, TGF-b1, TLR-4, F4/80 and TNF-a in kidneys
↓ BUN and UA
↑ CAT
[116]
Flavonoids and terpenoids[117]↓ blood urea and creatinine
↑ glutathione peroxidase activity
[117]
Myricetin[118]↓ BUN, β2-MG, and Cys C
↑ GR activity, TAS and GPx, MDA
↓ TNF-α and IL-1β
[118]
Tannins, phenolics, flavonoids, steroid glycosides, terpenoids, and saponins[119]↓ serum concentrations of BUN and creatinine
↓ renal tubular and glomerular alterations
[119]
Gallic acid, (+)-catechin, (–)-epicatechin, and ellagic acid[120]↓ ALT and AST activities
↓ MDA, APOP and NO in plasma, heart, and kidney
↓ uric acid level, and plasma creatinine level
[120]
Chlorogenic acid, diosmin, and caffeic acid[122]↓ urinary NGAL, endothelin-1, and clusterin levels
↓ BUN, creatinine, 8-OHda, isoprostane, adiponectin, and cystatin
↓ serum XOD, liver XOD, serum ADA, liver ADA, and serum UA levels
[121]
6–Shogaol, 6–Paradol, and 6–Gingerol[123]↓ plasma and blood creatinine and urea
↓ GSH level
↑ SOD level
[123]
Ferulic acid, caffeic acid and vanillic acid[124]↓ MDA, APOP, uric acid and creatinine levels in the blood, IL-1, IL-6, TNF-α, TGF-β1, and NF-κB
↑ catalase and SOD activities in the kidneys
[124]
Chlorogenic, vanillic, caffeic, syringic, p-coumaric, and ferulic acids[126]↓ serum urea, uric acid and creatinine levels
↑ kidney SOD, CAT, GPx, GST, and GR activities
[125]
Lutein and chlorophyll[127]↑ CAT and SOD
↓ urea, creatinine, uric acid, GGT, CK, BUN, and β2-microglobulin
↓ levels of TNFα, IL-6, IL-β, and TGF-α
[127]
Benzoic acids[128]↓ levels of Nrf2 and SOD1 in renal tissue
Normal architecture of renal tissue (renal corpuscle, proximal and distal convoluted tubules, and collecting ducts)
[128]
Rosmarinic acid[130]↓ serum creatinine, BUN, and uric acid[129]
Eugenol[131]↓ oxidative stress, inflammation, and apoptosis
↓ proinflammatory markers (IL6 and TNF-α)
[131]
Chlorogenic acid, hydroxytyrosol, and tyrosol[132]↓ creatinine, uric acids, and BUN[132]
Pyrogallol, 4-hydroxybenzoic acid, gallic acid, and ferulic acid [134]↑ GSH, SOD, CAT
↓ MDA
[133]
Secoisolariciresinol diglucoside[135]↓ levels of NO and MPO
↑ superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase
[135]
Proanthocyanidins ↑ SOD activity
↓ MDA content, CR and BUN levels
[136]
Kaempferol and quercetin derivatives ↓ serum UA level, liver MDA, serum Cr, and serum TG[138]
Kaempferol, luteolin, and apigenin [140]↓ serum UA, XOD, MCP-1, TNF-α, Cr, and BUN[139]
Pyrrolidide amides, chalcones, and flavonols[141]↑ urine output
↓ calcium oxalate crystallization
[141]
Retinol and caffeoylquinic acid[142]Ameliorated abnormal urinary levels of calcium, oxalate, phosphate, magnesium, citrate, protein, and uric acid
↓ serum BUN, creatinine, and uric acid levels
[142]
Narirutin, neohesperidin, hesperidin, rutin and citric acid[143]↓ nucleation and growth and aggregation of calcium oxalate crystals
↓ renal tubular dilation and renal tissue deterioration
[143]
Flavonoids, steroidal saponin, and organic acids[144]Urinary excretion of total protein, urea, creatinine, sodium, potassium, calcium, and chloride[144]
Gallic acid[145]↑ ACE inhibitory activity[145]
Increased (↑), decresead (↓), C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), pro-inflammatory cytokines (interleukin 6 (IL-6)), superoxide dismutase (SOD), malondialdehyde (MDA), total antioxidant capacity (TAC), tumor necrosis factor α (TNF-α), interleukin 1b (IL-1b), transforming growth factor b1 (TGF-b1), toll like receptor-4 (TLR-4), catalase (CAT), glutathione-S-transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GR), renal reduced glutathione (GSH).
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

Baptista, F.; Paié-Ribeiro, J.; Almeida, M.; Barros, A.N. Exploring the Role of Phenolic Compounds in Chronic Kidney Disease: A Systematic Review. Molecules 2024, 29, 2576. https://doi.org/10.3390/molecules29112576

AMA Style

Baptista F, Paié-Ribeiro J, Almeida M, Barros AN. Exploring the Role of Phenolic Compounds in Chronic Kidney Disease: A Systematic Review. Molecules. 2024; 29(11):2576. https://doi.org/10.3390/molecules29112576

Chicago/Turabian Style

Baptista, Filipa, Jessica Paié-Ribeiro, Mariana Almeida, and Ana Novo Barros. 2024. "Exploring the Role of Phenolic Compounds in Chronic Kidney Disease: A Systematic Review" Molecules 29, no. 11: 2576. https://doi.org/10.3390/molecules29112576

Article Metrics

Back to TopTop