Next Article in Journal
Comparative Analysis of Blood Transfusion Accuracy and Hemolysis Rate of Transfusion Cartridge Set Between Conventional Infusion Pumps and Cylinder-Type Infusion Pumps
Next Article in Special Issue
Effect of Comparable Carbon Chain Length Short- and Branched-Chain Fatty Acids on Adipokine Secretion from Normoxic and Hypoxic Lipopolysaccharide-Stimulated 3T3-L1 Adipocytes
Previous Article in Journal
Targeting Sepsis: Disease Tolerance, Immune Resilience, and Compartmentalized Immunity
Previous Article in Special Issue
Serum Adiponectin Is Elevated in Critically Ill Patients with Liver Disease and Associated with Decreased Overall Survival
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications

by
Ismael Valladolid-Acebes
The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-171 76 Stockholm, Sweden
Biomedicines 2024, 12(11), 2422; https://doi.org/10.3390/biomedicines12112422
Submission received: 13 September 2024 / Revised: 17 October 2024 / Accepted: 17 October 2024 / Published: 22 October 2024
(This article belongs to the Special Issue Recent Advances in Adipokines—2nd Edition)

Abstract

:
Background: Leptin, an adipokine essential for regulating energy balance, exerts important effects on brain function, notably within the hippocampus, a region integral to learning and memory. Leptin resistance, characterized by diminished responsiveness to elevated leptin levels, disrupts hippocampal function and exacerbates both obesity and cognitive impairments. Scope: This review critically examines how leptin resistance impairs hippocampal synaptic plasticity processes, specifically affecting long-term potentiation (LTP) and long-term depression (LTD), which are crucial for cognitive performance. Findings: Recent research highlights that leptin resistance disrupts N-methyl-D-aspartate (NMDA) receptor dynamics and hippocampal structure, leading to deficits in spatial learning and memory. Additionally, high-fat diets (HFDs), which contribute to leptin resistance, further deteriorate hippocampal function. Potential therapeutic strategies, including leptin sensitizers, show promise in mitigating brain disorders associated with leptin resistance. Complementary interventions such as caloric restriction and physical exercise also enhance leptin sensitivity and offer potential benefits to alleviating cognitive impairments. Aims of the review: This review synthesizes recent findings on the molecular pathways underlying leptin resistance and its impact on synaptic transmission and plasticity in the hippocampus. By identifying potential therapeutic targets, this work aims to provide an integrated approach for addressing cognitive deficits in obesity, ultimately improving the quality of life for affected individuals.

1. Introduction

Leptin, a 16 kDa polypeptide hormone encoded by the ob gene, is a key regulator of energy homeostasis with profound implications for metabolic functions [1,2]. First described in 1994 [1], the role of leptin extends beyond its foundational action in regulating appetite and energy expenditure. It acts as a critical neuroendocrine signal, influencing a broad spectrum of physiological processes through its interaction with an array of receptors distributed across nearly every tissue in the body [3,4,5,6,7,8,9,10,11].
The biological actions of leptin are linked to the leptin receptor (ObR), which manifests in multiple isoforms with varying functions [4,11,12,13,14]. The long isoform, or ObRb, is predominantly expressed in the hypothalamus and is crucial for mediating the anorexigenic effects of the hormone and integrating metabolic signals to influence food intake and energy expenditure [13,14,15,16,17]. Conversely, soluble isoforms like ObRe modulate leptin levels in the circulation, thereby buffering against excessive hormonal fluctuations [11,18,19,20,21,22]. The complex signaling cascades initiated by leptin binding—particularly through the Janus kinase/signal transducer and activator of transcription proteins (Jak/STATs) pathway, alongside the phosphoinositide 3-kinases (PI3Ks) and mitogen-activated protein kinase (MAPK) pathways—underscore its role in cellular processes such as metabolic regulation, synaptic plasticity and neuronal survival [4,6,23,24,25,26,27,28].
However, in the context of obesity and metabolic disorders, the physiological actions of leptin are compromised by the development of leptin resistance [2,29]. This condition, characterized by impaired leptin signaling despite elevated circulating levels, contributes to a paradoxical increase in body weight and metabolic dysregulation [29,30,31,32,33]. The persistence of leptin resistance in obesity and its impact on cognitive function, particularly within the hippocampus, highlights a critical intersection between metabolic and neurological health [34,35] (Figure 1).
The hippocampus, located in the medial temporal lobe, is vital for both spatial and declarative memory [36]. It processes information through a complex network of neurons organized in a distinct laminar structure, receiving input from various pathways, such as the perforant path, mossy fibers and Schaffer collaterals [36,37,38,39]. This structure is essential for encoding spatial information and consolidating short- and long-term memories [36]. Neuronal plasticity within the hippocampus, facilitated by mechanisms like long-term potentiation (LTP) and long-term depression (LTD), underpins learning and memory processes both in rodents and in humans [40].
Preclinical evidence suggests that leptin resistance impairs hippocampal synaptic plasticity and cognitive performance, exacerbating learning and memory deficits [41,42,43,44,45,46]. The effects of high-fat diets (HFDs) and other metabolic perturbations on leptin signaling in the mouse brain point to a broader impact on cognitive function and suggest potential therapeutic avenues (Figure 1). In-field research demonstrates the therapeutic potential of leptin analogs and sensitizers [47,48,49], which can be potentially beneficial for cognitive deficits in both rodents and humans, albeit complicated by the phenomenon of leptin resistance.
Despite significant advances in understanding the relationship between hippocampal leptin resistance and cognitive decline, several gaps in knowledge remain. Firstly, the exact molecular mechanisms linking leptin resistance to synaptic plasticity and cognition are unclear [40,41,42,43,44,45,46]. Additionally, the impact of leptin resistance at different life stages and its role in hippocampal function are poorly understood [46]. Finally, translational challenges from animal models to humans persist, and while therapies like leptin sensitizers show promise [47,48,49], their effectiveness in treating cognitive decline needs further research for clinical application.
This review seeks to offer a comprehensive examination of the influence of leptin resistance on hippocampal function, with particular emphasis on the mechanisms affecting glutamatergic transmission and synaptic plasticity in the context of obesity. By synthesizing recent and past findings on the molecular pathways involved in leptin resistance and their implications for cognitive function, this review aims to identify potential therapeutic targets to address cognitive deficits linked to leptin resistance in obesity.

2. Leptin: Biological Role and Actions

Leptin is a 167-amino acid polypeptide encoded by the ob gene [1]. It starts with a 2-amino acid signal sequence that is cleaved off before the protein enters the bloodstream. Consequently, the mature leptin molecule, with a molecular weight of 16 kDa, consists of 146 amino acids and resembles class I cytokines in its tertiary structure [1,50].
The biological activity of leptin hinges on a critical disulfide bond in its structure. Despite some interspecies variation, human leptin is a highly conserved protein; for example, sharing a homology of 84% with mouse leptin and 83% with rat leptin [1,2,51,52,53]. While predominantly produced in white adipose tissue [1,2,51,52,53,54,55], leptin is also synthesized by brown adipose tissue and various other organs, including the brain, skeletal muscles, stomach, breasts, ovaries and placenta (during pregnancy) [56,57,58,59,60,61,62,63,64,65].
Leptin synthesis is influenced by several factors. For instance, age, HFDs, insulin and glucocorticoids all enhance leptin production, while fasting, exercise, heat and adrenergic stimulation suppress it [62,66,67]. Its secretion follows a pulsatile pattern and is modulated by other hormones like insulin and cholecystokinin [67]. Additionally, leptin secretion exhibits a circadian rhythm, with peak levels occurring during the day in humans and at night in rodents [68,69,70].
Once in circulation, leptin can either be free or bound to plasma proteins [68]. Its plasma concentrations vary from around 5 ng/mL in individuals with a normal weight and can reach up to 25–100 ng/mL in those with obesity [2]. The hormone has a short half-life of 25 min in its endogenous form and 90 min in its exogenous form, with rapid clearance primarily through the kidneys [71].
Leptin is a key player in the neuroendocrine and behavioral regulation of food intake and energy balance [2,51,52,53,54,55]. It has an extensive range of effects, both centrally in the brain and peripherally across various tissues. This wide-ranging influence is largely attributed to its receptors, which are distributed throughout nearly every tissue in the body.

3. Leptin Receptors

The leptin receptor, first discovered in 1995, is a membrane-bound class I cytokine receptor [12]. Researchers have identified up to six isoforms of the leptin receptor: ObRa, ObRb, ObRc, ObRd, ObRe and ObRf. These isoforms all feature an N-terminal extracellular domain for leptin binding, a transmembrane domain of 34 amino acids anchoring the receptor to the cell membrane and a variable C-terminal intracellular domain [4,13,14]. The diversity among these isoforms leads to their classification into three categories: short isoforms (ObRa, ObRc, ObRd, ObRf) with 30–40 amino acids in the C-terminal region; the long isoform (ObRb) with a 302-amino acid C-terminal; and the soluble isoform (ObRe), which lacks this domain altogether [12,13,14].
The soluble isoform, ObRe, plays a crucial role in modulating leptin levels in the bloodstream by binding to circulating leptin and acting as a buffer [16,17,18,19,20]. In contrast, the long isoform ObRb, highly expressed in the hypothalamus, is central to the anorexigenic effects of leptin [12,13,14,15]. ObRb uniquely contains the intracellular domain required for leptin signal transduction, primarily through the Jak/STAT pathway [4,13,14,15,16,23].
Short isoforms of the leptin receptor are involved in recruiting Jak proteins and appear to play roles in leptin internalization, degradation and transport across the blood–brain barrier and the choroid plexus [13,24,26,27,28] (Figure 1). Leptin receptors are widely distributed throughout the body, with expression detected in various regions of the central nervous system (CNS), including the hypothalamus, cerebral cortex, ventral tegmental area, nucleus of the solitary tract and hippocampus [3,4,5,7,8,9,10]. Additionally, they are found in peripheral tissues such as the spleen, liver, skeletal muscle, heart and adipose tissue [6]. This widespread distribution highlights the pivotal role of leptin in regulating numerous physiological processes throughout the body.

3.1. Intracellular Signaling of Leptin Coupled to the Leptin Receptor

The Jak/STAT pathway is the primary intracellular signaling cascade of leptin coupled to ObRb. However, ObRb and, to a lesser extent, ObRa can also activate alternative pathways, such as the PI3K and the MAPK pathways [4,6,23,24,25,26,27,28] (Figure 2).

3.1.1. Jak/STAT Pathway

In the hypothalamus, the leptin-driven activation of STAT3 is closely linked to weight loss and increased energy expenditure in physiological conditions [2,17,23,52,66,72,73] (Figure 2). Activated or phosphorylated STAT3 acts as a pivotal transcription factor, orchestrating the expression of genes like suppressor of cytokine signaling 3 (SOCS3) and the tyrosine-protein phosphatase non-receptor type 1B (PTP1b), both of which in turn mediate crucial feedback inhibition in the leptin receptor [2,74] (Figure 2). Beyond the hypothalamus, in areas like the hippocampus, leptin influences neuronal survival through the Jak/STAT and PI3K pathways by enhancing the production of antioxidants, such as manganese superoxide dismutase and the anti-apoptotic protein Bcl-xL (B-cell lymphoma-extra-large) [75]. Additionally, leptin modulates hippocampal neuronal excitability by increasing intracellular Ca2+ concentrations in glutamatergic neurons induced by N-methyl-D-aspartate (NMDA) receptor facilitation [76] (Figure 2). This results in modifications to AMPA receptor trafficking, which in turn induces persistent changes in the excitatory synaptic strength within the hippocampus [77] (Figure 2). Rodents deficient in leptin signaling or receptors exhibit notable deficits in spatial learning and memory, highlighting the role of leptin in hippocampal synaptic plasticity and cognitive function [41,78,79,80].

3.1.2. PI3K/Akt Pathway

The PI3K pathway, shared by leptin and insulin signaling, is activated when leptin binds to its receptor, leading to the phosphorylation of insulin receptor substrates (IRS1/2) and the subsequent activation of protein kinase B (PKB/Akt) [6,25,26,27,28] (Figure 2). Akt regulates numerous physiological processes, including glucose metabolism, cell proliferation, apoptosis, transcription and migration [27,28]. In the hippocampus, Akt activation is crucial for synaptic plasticity, with studies showing that the PI3K/Akt pathway governs LTD through NMDA receptor synthesis in the CA1 region [81,82,83,84,85]. This underlines the need of a fully functional PI3K/Akt pathway for leptin-mediated synaptic plasticity within the hippocampus.

3.1.3. MAPK Pathway

Leptin also activates the MAPK pathway via its receptors [4,13,14]. Leptin binding prompts the phosphorylation of Tyr985, which facilitates the binding of extracellular signal-regulated kinases 1/2 (ERK1/2) and the activation of transcription factors like c-Fos and Erg-1 (early growth response protein 1), involved in cell proliferation and differentiation [27,28] (Figure 2). In the hippocampus, MAPK activation counters the effects of the PI3K/Akt pathway. Electrophysiological studies show that inhibiting MAPK prevents increased neuronal excitability [85,86,87,88], and in vitro research suggests complementary mechanisms between PI3K/Akt and MAPK pathways in regulating synaptic plasticity [89]. Notably, peak Erg-1 induction correlates with high MAPK activity and low phosphorylated Akt levels, indicating that the interplay between these pathways is crucial for hippocampal synaptic plasticity and cognitive processes [86,87,88,89,90,91].

4. Physiological Effects of Leptin

Leptin orchestrates both food intake and energy expenditure through complex central and peripheral mechanisms. In the hypothalamus, leptin exerts its effects by modulating the expression of neuropeptides (Figure 2). It is known that leptin decreases orexigenic peptides, like neuropeptide Y (NPY) and Agouti-related peptide (AgRP), while increasing anorexigenic peptides, such as pro-opiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART) [29,92,93,94,95,96] (Figure 2). Additionally, leptin enhances neurotrophin expression, including brain-derived neurotrophic factor (BDNF), in the ventromedial nucleus [97,98,99]. Disruptions in these pathways, along with hyperleptinemia and central leptin resistance, may contribute to reductions in neurogenesis, altered neuronal plasticity and cognitive deficits [97,98,99,100] (Figure 1).
Peripherally, leptin boosts energy expenditure and stimulates fatty acid β-oxidation in the mouse liver [101,102]. In adipose tissue, leptin administration promotes lipolysis in ob/ob mice but not in db/db mice lacking the leptin receptor [103]. In rat skeletal muscles, leptin enhances β-oxidation via AMPK activation and improves insulin sensitivity by reducing triglyceride accumulation [104]. Cardiac tissue from rats also benefits from leptin, which stimulates β-oxidation independently of AMPK and acts as a cardiotrophic factor, promoting cardiomyocyte growth [105,106].
Over the years, research has increasingly focused on the actions of leptin on neuronal functions beyond energy regulation [36]. Preclinical suggests that leptin influences excitatory synaptic transmission and plasticity in the hippocampus [36,76,80,85,87,88,107] (Figure 1 and Figure 2). Direct leptin administration in the dentate gyrus or the CA1 region of the hippocampus enhances LTP and spatial memory in rodents [108,109]. Moreover, acute leptin application in hippocampal slices facilitates NMDA receptor-dependent synaptic plasticity, converting short-term potentiation into LTP and enhancing its induction [76,110] (Figure 2). Leptin also induces depotentiation, a novel form of LTD dependent on NMDA receptor synthesis, which counters hyperexcitability and maintains synaptic balance [82,83] (Figure 2). Furthermore, leptin increases dendritic spine density and mobility, highlighting its role in regulating excitatory synaptic efficacy [111].
In clinically relevant disease models, dysfunctional leptin signaling has been implicated in a number of hippocampal impairments. For instance, genetically engineered mouse models lacking leptin signaling exhibit impaired hippocampal plasticity, including altered LTP and LTD, that correlates with spatial learning and memory deficits [41,79,112]. In murine models of diet-induced obesity (DIO), impaired hippocampal leptin signaling has been shown to compromise spatial learning and memory and disrupt hippocampal glutamate metabolism and neurochemistry [113,114]. Moreover, aberrant leptin signaling adversely affects synaptic plasticity within the hippocampus, creating alterations in LTD as well as changes in dendritic density and morphology [34,35,113,114]. Importantly, the detrimental impact of impaired leptin signaling on hippocampal function and cognitive performance observed in murine models parallels human data, thus validating the impact of dietary patterns and fat intake on brain morphology and cognitive health across the lifespan [115,116,117,118].

5. Hippocampal Leptin Resistance

Leptin resistance represents a pivotal pathophysiological aspect of obesity, characterized by an impairment in the ability of the CNS to respond to leptin [29] (Figure 1). Leptin typically functions to inhibit food intake and enhance energy expenditure, thereby facilitating weight loss [90,91,92,93,94] (Figure 2). However, in the state of leptin resistance, the response of the brain to leptin is compromised despite elevated circulating levels of the hormone, leading to an unremitting increase in body weight [2,29] (Figure 1).
In the context of obesity, an excessive intake of calories results in adipocyte hypertrophy and hyperplasia (Figure 1), which precipitates a state of chronic low-grade inflammation and the subsequent infiltration of immune cells within the adipose tissue [119,120,121]. This inflammatory milieu is associated with an augmented release of pro-inflammatory cytokines and a perturbation in the homeostasis of crucial adipokines, including leptin and adiponectin [119,120,121]. Empirical evidence derived from obesity models indicates that, despite elevated leptin concentrations, the anticipated physiological responses are not achieved due to the underlying resistance mechanisms [2,29] (Figure 1).
The molecular mechanisms of leptin resistance involve perturbations in critical signaling pathways, such as STAT3, PTP1b and SOCS3, as well as alterations in the leptin receptor [2,29,33,122,123,124,125,126]. These disruptions impede the effective transduction of leptin signals, thereby exacerbating the obesity phenotype.
Leptin resistance has major implications for hippocampal function and plasticity [41,79,112,114,115,116,117,118] (Figure 1). In physiological conditions, leptin facilitates synaptic plasticity by inducing LTP and modulating glutamatergic transmission [108,109] (Figure 2). However, in leptin-resistant conditions, such as those resulting from HFDs and age-related impairments, these actions are diminished or absent [34,35,113,114,115,116,117,118] (Figure 1). For example, the ability of leptin to induce LTP or promote LTD at CA1 synapses is reduced, if not completely lost, in the hippocampus of DIO and aged mice [114,115,116,117,118].
In animal models of leptin resistance, deficits in hippocampal-dependent behaviors are observed [41,42,43,44,45,46]. For instance, leptin-resistant rodents exhibit impaired spatial learning and memory in water maze tasks, despite normal locomotor activity [41,127]. Studies of genetic models like the Zucker rats demonstrate important cognitive impairments, particularly under conditions that challenge hippocampal plasticity, such as tasks requiring longer inter-trial intervals [42].
These changes suggest that leptin resistance disrupts normal hippocampal function by impairing synaptic plasticity, which underlies cognitive deficits [41,42,43,44,45,46] (Figure 1). Furthermore, leptin resistance often accompanies other metabolic disturbances that may compound these effects [41,42,43,44,45,46]. Thus, addressing leptin resistance and its impact on hippocampal function is crucial for understanding and mitigating cognitive impairments associated with metabolic disorders and aging.

6. Cognitive Consequences of Hippocampal Leptin Resistance

The consequences of hippocampal leptin resistance extend beyond metabolic dysregulation to encompass profound effects on learning, memory and reward processing, which in turn can exacerbate overeating and obesity in rodents and humans [36,41,42,43,44,45,46,128]. Emerging evidence shows that hippocampus-dependent mechanisms play a crucial role in the regulation of food intake by influencing higher-order cognitive processes that govern decision-making and impulse control [46,98,129,130,131,132]. The hippocampus, traditionally recognized for its role in learning and memory, also modulates energy homeostasis and eating behaviors, suggesting a critical intersection between cognitive function and metabolic control [46,98,129,130,131,132]. The disruption of these processes through DIO, particularly via the consumption of HFDs and palatable foods, not only leads to hippocampal dysfunction but also triggers addictive-like behaviors that reinforce overeating [133,134].
Research has shown that high-calorie diets adversely affect hippocampal structure and function, impairing synaptic plasticity and neurogenesis, which are essential for learning and memory [78,98,135,136,137,138]. This suggests that the hippocampus is highly sensitive to dietary composition, particularly during critical developmental periods when the brain is more vulnerable to external insults, including neurotoxic agents like saturated fats [46,139]. The link between DIO and cognitive decline, including memory loss and an increased risk of neurodegenerative diseases, points out the potential long-term consequences of poor dietary habits established during adolescence, a period marked by substantial synaptic pruning and shifts in glutamatergic transmission [114,140,141,142].
Particularly concerning is the impact of HFDs on NMDA receptor function, which plays a pivotal role in synaptic plasticity and cognitive function. Developmental changes in NMDA receptor subunits, notably the transition from NR2B to NR2A, are crucial for synaptogenesis and neuronal connectivity, yet the effects of HFDs on these receptors during adolescence, as compared to adulthood, remain underexplored [143,144,145,146]. Understanding these mechanisms is essential, as the NMDA receptor has been proposed as a therapeutic target for treating eating disorders associated with obesity [147,148,149]. The potential irreversibility of obesity-induced hippocampal damage highlights the urgency of early interventions to prevent cognitive impairments and to address the addictive nature of food that undermines efforts to control intake, contributing to the perpetuation of obesity [150].

7. Implications of Hippocampal Leptin Resistance for Executive Function and Alzheimer’s Disease

In rodent models and humans, obesity-induced leptin resistance in the hippocampus has significant implications for executive functions, including decision-making, impulse control and attention regulation, which are vital for maintaining cognitive control over food intake [131,151,152,153,154,155]. When leptin resistance occurs in the hippocampus, these cognitive processes are impaired, leading to weakened appetite regulation and reinforcing maladaptive eating behaviors [131,151,152,153,154,155]. This dysfunction can diminish working memory and executive function, making it challenging for individuals to prioritize long-term goals over immediate food-related impulses [131,151,152,153,154,155]. These derangements become even more pronounced in neurodegenerative conditions such as Alzheimer’s disease [85,116,156,157]. In patients with Alzheimer’s disease, leptin signaling in the hippocampus is decreased and leptin localization is shifted, being more abundant in reactive astrocytes and less reactive in neurons [158]. This deranged hippocampal leptin signaling can very well be a contributing factor to neuronal degeneration, linking metabolic dysfunction directly to cognitive decline in Alzheimer’s disease [158].
Both leptin resistance in obesity and disrupted leptin signaling in Alzheimer’s disease share a common pathway of cognitive impairment, particularly affecting memory and executive function both in rodents and humans [85,116,156,157,158]. This relationship underscores the potential for targeting leptin pathways as a therapeutic strategy to mitigate cognitive decline and address the metabolic aspects of neurodegenerative diseases [85,116,156,157,158]. By understanding these mechanisms, we can better explore interventions that may enhance leptin sensitivity and improve cognitive outcomes in affected individuals.

8. Therapeutic Interventions and Future Directions

Emerging evidence suggests that targeting leptin signaling could be a promising therapeutic strategy to mitigate the cognitive decline associated with hippocampal leptin resistance [159,160,161]. The therapeutic potential of leptin has been proven by studies demonstrating that supplementation with the hormone can reverse cognitive deficits in leptin-deficient models, in addition to its broader neurotrophic effects in regions such as the cerebellum and anterior cingulate gyrus [162,163,164].
In individuals with leptin deficiency, leptin replacement therapy not only normalizes metabolic abnormalities but also induces structural and functional improvements in key areas involved in cognitive and emotional processes, including the hippocampus [162,163,164,165,166]. Importantly, it has been shown that the exogenous administration of leptin improves neurogenesis, axonal growth, synaptogenesis, LTP and memory formation [77,167,168,169]. Therefore, restoring leptin signaling in the brain could be a viable approach to prevent or even reverse hippocampal atrophy and cognitive decline in patients with leptin deficiency.
However, the clinical application of leptin therapy presents significant challenges, particularly due to the phenomenon of leptin resistance in obesity [29]. In leptin-resistant individuals, elevated leptin levels fail to exert their normal physiological effects, reducing the efficacy of leptin therapy [29]. This is analogous to insulin resistance in type 2 diabetes, where high insulin levels do not produce the expected metabolic effects [170]. In this context, the development of leptin sensitizers, such as amylin analogs and Celastrol, which have shown promise in preclinical models [47,48,49], may offer a novel therapeutic avenue.
Recent clinical trials have explored various amylin analogs, including pramlintide, cagrilintide, davalintide (AC2307) and GUBamy (GUB014295), that have demonstrated potential in addressing overweight and/or obesity due to their actions as leptin sensitizers [47,171] (Table 1). These analogs might represent an important therapeutic venue in the future to overcome brain derangements associated with leptin resistance [172,173,174]. For instance, trials like NCT00690235, which focused on schizophrenia and medication-induced weight gain, and NCT03560960, which was aimed at Alzheimer’s disease and mild cognitive impairment, highlight the broader implications of improving leptin sensitivity in the context of brain dysfunction (Table 1). Thus, pramlintide appears to be a promising candidate for not only facilitating weight loss and enhancing glycemic control but also potentially offering cognitive benefits through improved metabolic function [172,173].
Similarly, cagrilintide, a novel amylin analog, has proven its safety and efficacy at reducing weight and improving metabolic outcomes in patients with overweight or obesity [175,176,177,178,179], and has a number of studies in phase III (Table 1). The relevance of these outcomes might extend to neurological conditions, where metabolic health plays a critical role in cognitive function [47,160,161]. For example, the potential role of cagrilintide in improving insulin sensitivity, reducing cardiovascular risk markers and regulating energy expenditure may offer therapeutic benefits in brain disorders exacerbated by metabolic impairments, such as Alzheimer’s disease [47,175,176,177,178,179].
In addition to amylin analogs, Celastrol, a plant-derived compound, has been investigated for its potential as a leptin sensitizer (Table 1). A clinical trial (NCT05494112) is currently underway to evaluate the efficacy of Celastrol in treating obesity by restoring leptin sensitivity [48] (Table 1), which may have important implications for neurodegenerative diseases in the future.
Preclinical studies suggest Celastrol enhances leptin signaling, potentially reversing leptin resistance, promoting weight loss and improving brain function in metabolic impairment contexts [48]. Although clinical data is still emerging, Celastrol represents a promising approach to overcoming the limitations of leptin therapy in resistant individuals [48], with potential benefits for both metabolic and cognitive health.
Collectively, these clinical trials highlight an increasing interest in utilizing leptin sensitizers to tackle not only obesity and metabolic disorders but also neurological conditions. This provides renewed hope for individuals suffering from cognitive impairments associated with leptin resistance and metabolic dysfunction.
In addition to pharmacological approaches, lifestyle interventions that modulate leptin sensitivity, such as caloric restriction and physical exercise, merit further investigation [180,181,182]. These interventions have been shown to enhance leptin signaling pathways [183,184] and may serve as adjunctive therapies to leptin sensitizers or replacement therapy.
It should, however, be acknowledged that the pleiotropic effects of leptin on neurotrophic factors, insulin and sex hormones complicate the interpretation of its direct effects on brain structure and function [26,185,186,187].
Future research should also focus on disentangling these complex interactions to better understand how leptin influences the hippocampus independently of its metabolic actions. This understanding is essential for developing targeted interventions that enhance leptin-dependent hippocampal function without exacerbating peripheral resistance to the hormone.
In summary, while the role of leptin in cognitive function is increasingly recognized, translating these findings into effective clinical therapies will require a nuanced approach. Addressing leptin resistance and optimizing leptin delivery to the brain represent critical future directions in the effort to combat cognitive decline and hippocampal dysfunction. Collaborative efforts integrating clinical trials, basic research and novel drug development will be essential to fully harness the therapeutic potential of leptin, its analogs and its sensitizers in neurodegenerative diseases.

9. Clinical Implications and Outlook

Hippocampal leptin resistance has relevant clinical implications for cognitive decline, particularly within the context of obesity and metabolic disorders [47,116,169,188] (Figure 1). The hippocampus, a critical region for learning, memory and decision-making, suffers when leptin signaling is disrupted, resulting in compromised synaptic plasticity and neurogenesis [34,36,44,98,131] (Figure 1). Such disruptions contribute to the cognitive deficits, including memory impairment and compromised spatial learning, frequently observed in individuals with leptin resistance [116,131,169,188,189] (Figure 1). Additionally, leptin resistance intensifies addictive-like behaviors associated with overeating, establishing a detrimental cycle that exacerbates obesity and further impairs hippocampal function [41,42,43,45,46,47,48,50,184,185,190,191]. Clinically, these findings underscore the unmet need for early intervention strategies targeting leptin resistance.
Potential approaches include pharmacological agents designed to enhance leptin sensitivity, such as amylin analogs or Celastrol, as well as lifestyle modifications like caloric restriction and increased physical activity [47,48,171,172,173,174,175,176,177,178,179,180,181,182,183,184] (Table 1). Addressing leptin resistance in the hippocampus may not only help mitigate cognitive decline but could also alleviate obesity-related comorbidities, including the risk of neurodegenerative diseases [188]. However, the complexity of the role of leptin in the CNS presents challenges [4,6,23,24,25,26,27], requiring further research to refine therapeutic strategies that enhance leptin signaling specifically within the brain without worsening peripheral leptin resistance. This emphasizes the critical importance of understanding and addressing hippocampal leptin resistance in the development of targeted, multifaceted treatments for cognitive health.

10. Conclusions

In conclusion, emerging understanding of hippocampal leptin resistance has revealed its profound impact on cognitive function [36,41,42,43,44,45,46,98,114,128,129,130,131,132,139,140,141,142,143,144,145,146,147,148,149,150], particularly in executive processes, such as decision-making, impulse control and memory [131,151,152,153,154,155]. This dysregulation not only exacerbates the challenges of obesity but also contributes to the pathophysiology of neurodegenerative diseases like Alzheimer’s disease [85,116,156,157]. As leptin signaling is compromised in these conditions, the link between metabolic dysfunction and cognitive decline becomes increasingly apparent [158].
Addressing hippocampal leptin resistance presents a promising avenue for therapeutic intervention [159,160,161]. Targeted strategies, including the development of leptin sensitizers and lifestyle modifications [47,48,171,172,173,174,175,176,177,178,179,180,181,182,183,184], may enhance cognitive outcomes while also mitigating the obesity epidemic [192]. These approaches hold the potential to restore synaptic plasticity and improve neurogenesis, critical components for maintaining cognitive health (Table 1, NCT03560960).
However, the complexity of the role of leptin in the central nervous system needs further investigation into the mechanisms underlying its effects [4,6,23,24,25,26,27]. Unraveling these pathways could lead to innovative treatments that specifically enhance leptin signaling within the brain, thereby alleviating cognitive deficits without exacerbating peripheral resistance [85,116,156,157,158].
Given the increasing prevalence of obesity and its associated cognitive impairments [156,192], early interventions targeting leptin resistance are essential. Ultimately, fostering an integrated understanding of the interplay between metabolism and cognition [158] will be crucial in developing effective strategies to improve brain health and quality of life for individuals affected by these conditions. The time is ripe for a multifaceted approach that bridges clinical research, pharmacotherapy and lifestyle interventions [47,48,171,172,173,174,175,176,177,178,179,180,181,182,183,184] to combat the deleterious effects of hippocampal leptin resistance and enhance cognitive resilience.

Funding

This research was funded by the Karolinska Institutet, the Alzheimer´s Foundation and the Sigurd and Elsa Goljes Foundation.

Data Availability Statement

The original data presented in Table 1 of this work are openly available in: https://clinicaltrials.gov/ (accessed on 16 October 2024).

Acknowledgments

Thank you very much to Montse Visa for the extraordinarily exciting scientific discussions within the field of this work.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432, Erratum in Nature 1995, 374, 479. [Google Scholar] [CrossRef] [PubMed]
  2. Friedman, J.M. Leptin and the endocrine control of energy balance. Nat. Metab. 2019, 1, 754–764. [Google Scholar] [CrossRef] [PubMed]
  3. Mercer, J.G.; Hoggard, N.; Williams, L.M.; Lawrence, C.B.; Hannah, L.T.; Trayhurn, P. Localization of leptin receptor mRNA and the long form splice variant (Ob-Rb) in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS Lett. 1996, 387, 113–116. [Google Scholar] [CrossRef] [PubMed]
  4. Bjørbaek, C.; Uotani, S.; da Silva, B.; Flier, J.S. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J. Biol. Chem. 1997, 272, 32686–32695. [Google Scholar] [CrossRef]
  5. Shioda, S.; Funahashi, H.; Nakajo, S.; Yada, T.; Maruta, O.; Nakai, Y. Immunohistochemical localization of leptin receptor in the rat brain. Neurosci. Lett. 1998, 243, 41–44. [Google Scholar] [CrossRef]
  6. Muoio, D.M.; Lynis Dohm, G. Peripheral metabolic actions of leptin. Best Pract. Res. Clin. Endocrinol. Metab. 2002, 16, 653–666. [Google Scholar] [CrossRef]
  7. Huo, L.; Grill, H.J.; Bjørbaek, C. Divergent regulation of proopiomelanocortin neurons by leptin in the nucleus of the solitary tract and in the arcuate hypothalamic nucleus. Diabetes 2006, 55, 567–573. [Google Scholar] [CrossRef]
  8. Fulton, S.; Pissios, P.; Manchon, R.P.; Stiles, L.; Frank, L.; Pothos, E.N.; Maratos-Flier, E.; Flier, J.S. Leptin regulation of the mesoaccumbens dopamine pathway. Neuron 2006, 51, 811–822. [Google Scholar] [CrossRef]
  9. Walker, C.D.; Long, H.; Williams, S.; Richard, D. Long-lasting effects of elevated neonatal leptin on rat hippocampal function, synaptic proteins and NMDA receptor subunits. J. Neurosci. Res. 2007, 85, 816–828. [Google Scholar] [CrossRef]
  10. Bjørbaek, C. Central leptin receptor action and resistance in obesity. J. Investig. Med. 2009, 57, 789–794. [Google Scholar] [CrossRef]
  11. Casado, M.E.; Collado-Pérez, R.; Frago, L.M.; Barrios, V. Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies. Int. J. Mol. Sci. 2023, 24, 1422. [Google Scholar] [CrossRef] [PubMed]
  12. Tartaglia, L.A.; Dembski, M.; Weng, X.; Deng, N.; Culpepper, J.; Devos, R.; Richards, G.J.; Campfield, L.A.; Clark, F.T.; Deeds, J.; et al. Identification and expression cloning of a leptin receptor, OB-R. Cell 1995, 83, 1263–1271. [Google Scholar] [CrossRef] [PubMed]
  13. Gorska, E.; Popko, K.; Stelmaszczyk-Emmel, A.; Ciepiela, O.; Kucharska, A.; Wasik, M. Leptin receptors. Eur. J. Med. Res. 2010, 15 (Suppl. S2), 50–54. [Google Scholar] [CrossRef]
  14. Obradovic, M.; Sudar-Milovanovic, E.; Soskic, S.; Essack, M.; Arya, S.; Stewart, A.J.; Gojobori, T.; Isenovic, E.R. Leptin and Obesity: Role and Clinical Implication. Front. Endocrinol. 2021, 12, 585887. [Google Scholar] [CrossRef]
  15. Schwartz, M.W.; Seeley, R.J.; Campfield, L.A.; Burn, P.; Baskin, D.G. Identification of targets of leptin action in rat hypothalamus. J. Clin. Investig. 1996, 98, 1101–1106. [Google Scholar] [CrossRef] [PubMed]
  16. Couce, M.E.; Burguera, B.; Parisi, J.E.; Jensen, M.D.; Lloyd, R.V. Localization of leptin receptor in the human brain. Neuroendocrinology 1997, 66, 145–150. [Google Scholar] [CrossRef]
  17. Boyle, C.A.; Kola, P.K.; Oraegbuna, C.S.; Lei, S. Leptin excites basolateral amygdala principal neurons and reduces food intake by LepRb-JAK2-PI3K-dependent depression of GIRK channels. J. Cell. Physiol. 2024, 239, e31117. [Google Scholar] [CrossRef]
  18. Löllmann, B.; Grüninger, S.; Stricker-Krongrad, A.; Chiesi, M. Detection and quantification of the leptin receptor splice variants Ob-Ra, b, and, e in different mouse tissues. Biochem. Biophys. Res. Commun. 1997, 238, 648–652, Erratum in Biochem. Biophys. Res. Commun. 1997, 241, 803. [Google Scholar] [CrossRef]
  19. Lammert, A.; Kiess, W.; Bottner, A.; Glasow, A.; Kratzsch, J. Soluble leptin receptor represents the main leptin binding activity in human blood. Biochem. Biophys. Res. Commun. 2001, 283, 982–988. [Google Scholar] [CrossRef]
  20. Chan, J.L.; Blüher, S.; Yiannakouris, N.; Suchard, M.A.; Kratzsch, J.; Mantzoros, C.S. Regulation of circulating soluble leptin receptor levels by gender, adiposity, sex steroids, and leptin: Observational and interventional studies in humans. Diabetes 2002, 51, 2105–2112. [Google Scholar] [CrossRef]
  21. Weinland, C.; Tanovska, P.; Kornhuber, J.; Mühle, C.; Lenz, B. Serum lipids, leptin, and soluble leptin receptor in alcohol dependence: A cross-sectional and longitudinal study. Drug Alcohol Depend. 2020, 209, 107898. [Google Scholar] [CrossRef] [PubMed]
  22. Bouna-Pyrrou, P.; Muehle, C.; Kornhuber, J.; Weinland, C.; Lenz, B. Body mass index and serum levels of soluble leptin receptor are sex-specifically related to alcohol binge drinking behavior. Psychoneuroendocrinology 2021, 127, 105179. [Google Scholar] [CrossRef] [PubMed]
  23. Vaisse, C.; Halaas, J.L.; Horvath, C.M.; Darnell, J.E., Jr.; Stoffel, M.; Friedman, J.M. Leptin activation of Stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice. Nat. Genet. 1996, 14, 95–97. [Google Scholar] [CrossRef]
  24. Uotani, S.; Bjørbaek, C.; Tornøe, J.; Flier, J.S. Functional properties of leptin receptor isoforms: Internalization and degradation of leptin and ligand-induced receptor downregulation. Diabetes 1999, 48, 279–286. [Google Scholar] [CrossRef] [PubMed]
  25. Banks, A.S.; Davis, S.M.; Bates, S.H.; Myers, M.G., Jr. Activation of downstream signals by the long form of the leptin receptor. J. Biol. Chem. 2000, 275, 14563–14572. [Google Scholar] [CrossRef]
  26. Bjørbaek, C.; Kahn, B.B. Leptin signaling in the central nervous system and the periphery. Recent Prog. Horm. Res. 2004, 59, 305–331. [Google Scholar] [CrossRef]
  27. Frühbeck, G. Intracellular signalling pathways activated by leptin. Biochem. J. 2006, 393 Pt 1, 7–20. [Google Scholar] [CrossRef]
  28. Saxton, R.A.; Caveney, N.A.; Moya-Garzon, M.D.; Householder, K.D.; Rodriguez, G.E.; Burdsall, K.A.; Long, J.Z.; Garcia, K.C. Structural insights into the mechanism of leptin receptor activation. Nat. Commun. 2023, 14, 1797. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, J.; Lai, F.; Hou, Y.; Zheng, R. Leptin signaling and leptin resistance. Med. Rev. 2022, 2, 363–384. [Google Scholar] [CrossRef]
  30. Bjørbaek, C.; Elmquist, J.K.; Frantz, J.D.; Shoelson, S.E.; Flier, J.S. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol. Cell 1998, 1, 619–625. [Google Scholar] [CrossRef]
  31. Barsh, G.S.; Farooqi, I.S.; O’Rahilly, S. Genetics of body-weight regulation. Nature 2000, 404, 644–651. [Google Scholar] [CrossRef] [PubMed]
  32. Reed, A.S.; Unger, E.K.; Olofsson, L.E.; Piper, M.L.; Myers, M.G., Jr.; Xu, A.W. Functional role of suppressor of cytokine signaling 3 upregulation in hypothalamic leptin resistance and long-term energy homeostasis. Diabetes 2010, 59, 894–906. [Google Scholar] [CrossRef] [PubMed]
  33. Wunderlich, C.M.; Hövelmeyer, N.; Wunderlich, F.T. Mechanisms of chronic JAK-STAT3-SOCS3 signaling in obesity. JAKSTAT 2013, 2, e23878. [Google Scholar] [CrossRef] [PubMed]
  34. Abbott, K.N.; Arnott, C.K.; Westbrook, R.F.; Tran, D.M.D. The effect of high fat, high sugar, and combined high fat-high sugar diets on spatial learning and memory in rodents: A meta-analysis. Neurosci. Biobehav. Rev. 2019, 107, 399–421. [Google Scholar] [CrossRef] [PubMed]
  35. Bakoyiannis, I.; Ducourneau, E.G.; N’diaye, M.; Fermigier, A.; Ducroix-Crepy, C.; Bosch-Bouju, C.; Coutureau, E.; Trifilieff, P.; Ferreira, G. Obesogenic diet induces circuit-specific memory deficits in mice. eLife 2024, 13, e80388. [Google Scholar] [CrossRef]
  36. Madison, F.N.; Bingman, V.P.; Smulders, T.V.; Lattin, C.R. A bird’s eye view of the hippocampus beyond space: Behavioral, neuroanatomical, and neuroendocrine perspectives. Horm. Behav. 2024, 157, 105451. [Google Scholar] [CrossRef]
  37. Witter, M.P. The perforant path: Projections from the entorhinal cortex to the dentate gyrus. Prog. Brain Res. 2007, 163, 43–61. [Google Scholar]
  38. Kobayashi, K. Hippocampal mossy fiber synaptic transmission and its modulation. Vitam. Horm. 2010, 82, 65–85. [Google Scholar]
  39. Szirmai, I.; Buzsáki, G.; Kamondi, A. 120 years of hippocampal Schaffer collaterals. Hippocampus 2012, 22, 1508–1516. [Google Scholar] [CrossRef]
  40. Bin Ibrahim, M.Z.; Benoy, A.; Sajikumar, S. Long-term plasticity in the hippocampus: Maintaining within and ‘tagging’ between synapses. FEBS J. 2022, 289, 2176–2201. [Google Scholar] [CrossRef]
  41. Li, X.L.; Aou, S.; Oomura, Y.; Hori, N.; Fukunaga, K.; Hori, T. Impairment of long-term potentiation and spatial memory in leptin receptor-deficient rodents. Neuroscience 2002, 113, 607–615. [Google Scholar] [CrossRef] [PubMed]
  42. Winocur, G.; Greenwood, C.E.; Piroli, G.G.; Grillo, C.A.; Reznikov, L.R.; Reagan, L.P.; McEwen, B.S. Memory impairment in obese Zucker rats: An investigation of cognitive function in an animal model of insulin resistance and obesity. Behav. Neurosci. 2005, 119, 1389–1395. [Google Scholar] [CrossRef] [PubMed]
  43. Stranahan, A.M.; Arumugam, T.V.; Cutler, R.G.; Lee, K.; Egan, J.M.; Mattson, M.P. Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nat. Neurosci. 2008, 11, 309–317. [Google Scholar] [CrossRef] [PubMed]
  44. Valladolid-Acebes, I.; Stucchi, P.; Cano, V.; Fernández-Alfonso, M.S.; Merino, B.; Gil-Ortega, M.; Fole, A.; Morales, L.; Ruiz-Gayo, M.; Del Olmo, N. High-fat diets impair spatial learning in the radial-arm maze in mice. Neurobiol. Learn. Mem. 2011, 95, 80–85. [Google Scholar] [CrossRef]
  45. Kamal, A.; Ramakers, G.M.; Gispen, W.H.; Biessels, G.J. Hyperinsulinemia in rats causes impairment of spatial memory and learning with defects in hippocampal synaptic plasticity by involvement of postsynaptic mechanisms. Exp. Brain Res. 2013, 226, 45–51, Erratum in Exp. Brain Res. 2013, 227, 421. [Google Scholar] [CrossRef]
  46. Valladolid-Acebes, I.; Fole, A.; Martín, M.; Morales, L.; Cano, M.V.; Ruiz-Gayo, M.; Del Olmo, N. Spatial memory impairment and changes in hippocampal morphology are triggered by high-fat diets in adolescent mice. Is there a role of leptin? Neurobiol. Learn. Mem. 2013, 106, 18–25. [Google Scholar] [CrossRef]
  47. Roth, J.D.; Roland, B.L.; Cole, R.L.; Trevaskis, J.L.; Weyer, C.; Koda, J.E.; Anderson, C.M.; Parkes, D.G.; Baron, A.D. Leptin responsiveness restored by amylin agonism in diet-induced obesity: Evidence from nonclinical and clinical studies. Proc. Natl. Acad. Sci. USA 2008, 105, 7257–7262. [Google Scholar] [CrossRef]
  48. Liu, J.; Lee, J.; Salazar Hernandez, M.A.; Mazitschek, R.; Ozcan, U. Treatment of obesity with celastrol. Cell 2015, 161, 999–1011. [Google Scholar] [CrossRef]
  49. Boyle, C.N.; Zheng, Y.; Lutz, T.A. Mediators of Amylin Action in Metabolic Control. J. Clin. Med. 2022, 11, 2207. [Google Scholar] [CrossRef]
  50. Madej, T.; Boguski, M.S.; Bryant, S.H. Threading analysis suggests that the obese gene product may be a helical cytokine. FEBS Lett. 1995, 373, 13–18. [Google Scholar] [CrossRef]
  51. Caro, J.F.; Sinha, M.K.; Kolaczynski, J.W.; Zhang, P.L.; Considine, R.V. Leptin: The tale of an obesity gene. Diabetes 1996, 45, 1455–1462. [Google Scholar] [CrossRef] [PubMed]
  52. Considine, R.V.; Sinha, M.K.; Heiman, M.L.; Kriauciunas, A.; Stephens, T.W.; Nyce, M.R.; Ohannesian, J.P.; Marco, C.C.; McKee, L.J.; Bauer, T.L.; et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 1996, 334, 292–295. [Google Scholar] [CrossRef] [PubMed]
  53. Fan, X.; Qin, R.; Yuan, W.; Fan, J.S.; Huang, W.; Lin, Z. The solution structure of human leptin reveals a conformational plasticity important for receptor recognition. Structure 2024, 32, 18–23.e2. [Google Scholar] [CrossRef]
  54. Drougard, A.; Fournel, A.; Valet, P.; Knauf, C. Impact of hypothalamic reactive oxygen species in the regulation of energy metabolism and food intake. Front. Neurosci. 2015, 9, 56. [Google Scholar] [CrossRef]
  55. Coccurello, R.; Maccarrone, M. Hedonic Eating and the “Delicious Circle”: From Lipid-Derived Mediators to Brain Dopamine and Back. Front. Neurosci. 2018, 12, 271. [Google Scholar] [CrossRef]
  56. Wiesner, G.; Vaz, M.; Collier, G.; Seals, D.; Kaye, D.; Jennings, G.; Lambert, G.; Wilkinson, D.; Esler, M. Leptin is released from the human brain: Influence of adiposity and gender. J. Clin. Endocrinol. Metab. 1999, 84, 2270–2274. [Google Scholar] [CrossRef]
  57. Ahima, R.S.; Flier, J.S. Leptin. Annu. Rev. Physiol. 2000, 62, 413–437. [Google Scholar] [CrossRef] [PubMed]
  58. Cinti, S. The adipose organ: Morphological perspectives of adipose tissues. Proc. Nutr. Soc. 2001, 60, 319–328. [Google Scholar] [CrossRef]
  59. Considine, R.V. Regulation of leptin production. Rev. Endocr. Metab. Disord. 2001, 2, 357–363. [Google Scholar] [CrossRef]
  60. Park, H.K.; Ahima, R.S. Physiology of leptin: Energy homeostasis, neuroendocrine function and metabolism. Metabolism 2015, 64, 24–34. [Google Scholar] [CrossRef] [PubMed]
  61. Ramos-Lobo, A.M.; Donato, J., Jr. The role of leptin in health and disease. Temperature 2017, 4, 258–291. [Google Scholar] [CrossRef] [PubMed]
  62. Caron, A.; Lee, S.; Elmquist, J.K.; Gautron, L. Leptin and brain-adipose crosstalks. Nat. Rev. Neurosci. 2018, 19, 153–165. [Google Scholar] [CrossRef] [PubMed]
  63. Lynes, M.D.; Tseng, Y.H. Deciphering adipose tissue heterogeneity. Ann. N. Y. Acad. Sci. 2018, 1411, 5–20. [Google Scholar] [CrossRef] [PubMed]
  64. Cinti, S. Adipose Organ Development and Remodeling. Compr. Physiol. 2018, 8, 1357–1431. [Google Scholar] [PubMed]
  65. Picó, C.; Palou, M.; Pomar, C.A.; Rodríguez, A.M.; Palou, A. Leptin as a key regulator of the adipose organ. Rev. Endocr. Metab. Disord. 2022, 23, 13–30. [Google Scholar] [CrossRef]
  66. Sasaki, T. Age-Associated Weight Gain, Leptin, and SIRT1: A Possible Role for Hypothalamic SIRT1 in the Prevention of Weight Gain and Aging through Modulation of Leptin Sensitivity. Front. Endocrinol. 2015, 6, 109. [Google Scholar] [CrossRef]
  67. Zhao, S.; Kusminski, C.M.; Elmquist, J.K.; Scherer, P.E. Leptin: Less Is More. Diabetes 2020, 69, 823–829. [Google Scholar] [CrossRef]
  68. Sinha, M.K.; Sturis, J.; Ohannesian, J.; Magosin, S.; Stephens, T.; Heiman, M.L.; Polonsky, K.S.; Caro, J.F. Ultradian oscillations of leptin secretion in humans. Biochem. Biophys. Res. Commun. 1996, 228, 733–738. [Google Scholar] [CrossRef]
  69. Sukumaran, S.; Xue, B.; Jusko, W.J.; Dubois, D.C.; Almon, R.R. Circadian variations in gene expression in rat abdominal adipose tissue and relationship to physiology. Physiol. Genom. 2010, 42, 141–152. [Google Scholar] [CrossRef]
  70. Ansarin, A.; Mahdavi, A.M.; Javadivala, Z.; Shanehbandi, D.; Zarredar, H.; Ansarin, K. The cross-talk between leptin and circadian rhythm signaling proteins in physiological processes: A systematic review. Mol. Biol. Rep. 2023, 50, 10427–10443. [Google Scholar] [CrossRef]
  71. Cumin, F.; Baum, H.P.; Levens, N. Leptin is cleared from the circulation primarily by the kidney. Int. J. Obes. Relat. Metab. Disord. 1996, 20, 1120–1126. [Google Scholar] [PubMed]
  72. Gao, Q.; Wolfgang, M.J.; Neschen, S.; Morino, K.; Horvath, T.L.; Shulman, G.I.; Fu, X.Y. Disruption of neural signal transducer and activator of transcription 3 causes obesity, diabetes, infertility, and thermal dysregulation. Proc. Natl. Acad. Sci. USA 2004, 101, 4661–4666. [Google Scholar] [CrossRef] [PubMed]
  73. Harris, R.B.S. Phosphorylation of STAT3 in hypothalamic nuclei is stimulated by lower doses of leptin than are needed to inhibit food intake. Am. J. Physiol. Endocrinol. Metab. 2021, 321, E190–E201. [Google Scholar] [CrossRef] [PubMed]
  74. Flak, J.N.; Myers, M.G., Jr. Minireview: CNS Mechanisms of Leptin Action. Mol. Endocrinol. 2016, 30, 3–12. [Google Scholar] [CrossRef] [PubMed]
  75. Guo, Z.; Jiang, H.; Xu, X.; Duan, W.; Mattson, M.P. Leptin-mediated cell survival signaling in hippocampal neurons mediated by JAK STAT3 and mitochondrial stabilization. J. Biol. Chem. 2008, 283, 1754–1763. [Google Scholar] [CrossRef]
  76. Shanley, L.J.; Irving, A.J.; Harvey, J. Leptin enhances NMDA receptor function and modulates hippocampal synaptic plasticity. J. Neurosci. 2001, 21, RC186. [Google Scholar] [CrossRef]
  77. McGregor, G.; Harvey, J. Leptin Regulation of Synaptic Function at Hippocampal TA-CA1 and SC-CA1 Synapses: Implications for Health and Disease. Neurochem. Res. 2019, 44, 650–660. [Google Scholar] [CrossRef]
  78. Farr, S.A.; Yamada, K.A.; Butterfield, D.A.; Abdul, H.M.; Xu, L.; Miller, N.E.; Banks, W.A.; Morley, J.E. Obesity and hypertriglyceridemia produce cognitive impairment. Endocrinology 2008, 149, 2628–2636. [Google Scholar] [CrossRef]
  79. Watanabe, T.; Sakamoto, K. Meta-analysis of cognitive and behavioral tests in leptin- and leptin receptor-deficient mice. Neurosci. Res. 2021, 170, 217–235. [Google Scholar] [CrossRef]
  80. Bonds, J.A.; Shetti, A.; Stephen, T.K.L.; Bonini, M.G.; Minshall, R.D.; Lazarov, O. Deficits in hippocampal neurogenesis in obesity-dependent and -independent type-2 diabetes mellitus mouse models. Sci. Rep. 2020, 10, 16368. [Google Scholar] [CrossRef]
  81. Hou, L.; Klann, E. Activation of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway is required for metabotropic glutamate receptor-dependent long-term depression. J. Neurosci. 2004, 24, 6352–6361. [Google Scholar] [CrossRef] [PubMed]
  82. Durakoglugil, M.; Irving, A.J.; Harvey, J. Leptin induces a novel form of NMDA receptor-dependent long-term depression. J. Neurochem. 2005, 95, 396–405. [Google Scholar] [CrossRef] [PubMed]
  83. Moult, P.R.; Harvey, J. Regulation of glutamate receptor trafficking by leptin. Biochem. Soc. Trans. 2009, 37 Pt 6, 1364–1368. [Google Scholar] [CrossRef]
  84. Zhao, F.; Siu, J.J.; Huang, W.; Askwith, C.; Cao, L. Insulin Modulates Excitatory Synaptic Transmission and Synaptic Plasticity in the Mouse Hippocampus. Neuroscience 2019, 411, 237–254. [Google Scholar] [CrossRef] [PubMed]
  85. Kommaddi, R.P.; Gowaikar, R.; Haseena, P.A.; Diwakar, L.; Singh, K.; Mondal, A. Akt activation ameliorates deficits in hippocampal-dependent memory and activity-dependent synaptic protein synthesis in an Alzheimer’s disease mouse model. J. Biol. Chem. 2024, 300, 105619. [Google Scholar] [CrossRef]
  86. Rosenkranz, J.A.; Frick, A.; Johnston, D. Kinase-dependent modification of dendritic excitability after long-term potentiation. J. Physiol. 2009, 587, 115–125. [Google Scholar] [CrossRef]
  87. Zhang, F.; Chen, J. Leptin protects hippocampal CA1 neurons against ischemic injury. J. Neurochem. 2008, 107, 578–587. [Google Scholar] [CrossRef]
  88. Guimond, D.; Diabira, D.; Porcher, C.; Bader, F.; Ferrand, N.; Zhu, M.; Appleyard, S.M.; Wayman, G.A.; Gaiarsa, J.L. Leptin potentiates GABAergic synaptic transmission in the developing rodent hippocampus. Front. Cell. Neurosci. 2014, 8, 235. [Google Scholar] [CrossRef] [PubMed]
  89. Bland, T.; Sahin, G.S.; Zhu, M.; Dillon, C.; Impey, S.; Appleyard, S.M.; Wayman, G.A. USP8 Deubiquitinates the Leptin Receptor and Is Necessary for Leptin-Mediated Synapse Formation. Endocrinology 2019, 160, 1982–1998. [Google Scholar] [CrossRef]
  90. Benz, A.H.; Shajari, M.; Peruzki, N.; Dehghani, F.; Maronde, E. Early growth response-1 induction by fibroblast growth factor-1 via increase of mitogen-activated protein kinase and inhibition of protein kinase B in hippocampal neurons. Br. J. Pharmacol. 2010, 160, 1621–1630. [Google Scholar] [CrossRef]
  91. Mishra, P.; Narayanan, R. Stable continual learning through structured multiscale plasticity manifolds. Curr. Opin. Neurobiol. 2021, 70, 51–63. [Google Scholar] [CrossRef]
  92. Stephens, T.W.; Basinski, M.; Bristow, P.K.; Bue-Valleskey, J.M.; Burgett, S.G.; Craft, L.; Hale, J.; Hoffmann, J.; Hsiung, H.M.; Kriauciunas, A.; et al. The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 1995, 377, 530–532. [Google Scholar] [CrossRef] [PubMed]
  93. Mizuno, T.M.; Mobbs, C.V. Hypothalamic agouti-related protein messenger ribonucleic acid is inhibited by leptin and stimulated by fasting. Endocrinology 1999, 140, 814–817. [Google Scholar] [CrossRef] [PubMed]
  94. Cowley, M.A.; Smart, J.L.; Rubinstein, M.; Cerdán, M.G.; Diano, S.; Horvath, T.L.; Cone, R.D.; Low, M.J. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 2001, 411, 480–484. [Google Scholar] [CrossRef] [PubMed]
  95. Gali Ramamoorthy, T.; Begum, G.; Harno, E.; White, A. Developmental programming of hypothalamic neuronal circuits: Impact on energy balance control. Front. Neurosci. 2015, 9, 126. [Google Scholar] [CrossRef]
  96. Timper, K.; Brüning, J.C. Hypothalamic circuits regulating appetite and energy homeostasis: Pathways to obesity. Dis. Model. Mech. 2017, 10, 679–689. [Google Scholar] [CrossRef]
  97. Ameroso, D.; Meng, A.; Chen, S.; Felsted, J.; Dulla, C.G.; Rios, M. Astrocytic BDNF signaling within the ventromedial hypothalamus regulates energy homeostasis. Nat. Metab. 2022, 4, 627–643. [Google Scholar] [CrossRef]
  98. Kanoski, S.E.; Meisel, R.L.; Mullins, A.J.; Davidson, T.L. The effects of energy-rich diets on discrimination reversal learning and on BDNF in the hippocampus and prefrontal cortex of the rat. Behav. Brain Res. 2007, 182, 57–66. [Google Scholar] [CrossRef]
  99. Park, H.R.; Park, M.; Choi, J.; Park, K.Y.; Chung, H.Y.; Lee, J. A high-fat diet impairs neurogenesis: Involvement of lipid peroxidation and brain-derived neurotrophic factor. Neurosci. Lett. 2010, 482, 235–239. [Google Scholar] [CrossRef] [PubMed]
  100. Hoane, M.R.; Swan, A.A.; Heck, S.E. The effects of a high-fat sucrose diet on functional outcome following cortical contusion injury in the rat. Behav. Brain Res. 2011, 223, 119–124. [Google Scholar] [CrossRef]
  101. Scarpace, P.J.; Matheny, M.; Pollock, B.H.; Tümer, N. Leptin increases uncoupling protein expression and energy expenditure. Am. J. Physiol. 1997, 273 Pt 1, E226–E230. [Google Scholar] [CrossRef] [PubMed]
  102. Liang, C.P.; Tall, A.R. Transcriptional profiling reveals global defects in energy metabolism, lipoprotein, and bile acid synthesis and transport with reversal by leptin treatment in ob/ob mouse liver. J. Biol. Chem. 2001, 276, 49066–49076. [Google Scholar] [CrossRef]
  103. Frühbeck, G.; Aguado, M.; Gómez-Ambrosi, J.; Martínez, J.A. Lipolytic effect of in vivo leptin administration on adipocytes of lean and ob/ob mice, but not db/db mice. Biochem. Biophys. Res. Commun. 1998, 250, 99–102. [Google Scholar] [CrossRef]
  104. Dube, J.J.; Bhatt, B.A.; Dedousis, N.; Bonen, A.; O’Doherty, R.M. Leptin, skeletal muscle lipids, and lipid-induced insulin resistance. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R642–R650. [Google Scholar] [CrossRef] [PubMed]
  105. Atkinson, L.L.; Fischer, M.A.; Lopaschuk, G.D. Leptin activates cardiac fatty acid oxidation independent of changes in the AMP-activated protein kinase-acetyl-CoA carboxylase-malonyl-CoA axis. J. Biol. Chem. 2002, 277, 29424–29430. [Google Scholar] [CrossRef] [PubMed]
  106. Rajapurohitam, V.; Gan, X.T.; Kirshenbaum, L.A.; Karmazyn, M. The obesity-associated peptide leptin induces hypertrophy in neonatal rat ventricular myocytes. Circ. Res. 2003, 93, 277–279. [Google Scholar] [CrossRef]
  107. Kanoski, S.E.; Hayes, M.R.; Greenwald, H.S.; Fortin, S.M.; Gianessi, C.A.; Gilbert, J.R.; Grill, H.J. Hippocampal leptin signaling reduces food intake and modulates food-related memory processing. Neuropsychopharmacology 2011, 36, 1859–1870. [Google Scholar] [CrossRef]
  108. Wayner, M.J.; Armstrong, D.L.; Phelix, C.F.; Oomura, Y. Orexin-A (Hypocretin-1) and leptin enhance LTP in the dentate gyrus of rats in vivo. Peptides 2004, 25, 991–996. [Google Scholar] [CrossRef]
  109. Farr, S.A.; Banks, W.A.; Morley, J.E. Effects of leptin on memory processing. Peptides 2006, 27, 1420–1425. [Google Scholar] [CrossRef]
  110. Oomura, Y.; Hori, N.; Shiraishi, T.; Fukunaga, K.; Takeda, H.; Tsuji, M.; Matsumiya, T.; Ishibashi, M.; Aou, S.; Li, X.L.; et al. Leptin facilitates learning and memory performance and enhances hippocampal CA1 long-term potentiation and CaMK II phosphorylation in rats. Peptides 2006, 27, 2738–2749. [Google Scholar] [CrossRef]
  111. O’Malley, D.; MacDonald, N.; Mizielinska, S.; Connolly, C.N.; Irving, A.J.; Harvey, J. Leptin promotes rapid dynamic changes in hippocampal dendritic morphology. Mol. Cell. Neurosci. 2007, 35, 559–572. [Google Scholar] [CrossRef] [PubMed]
  112. Porter, W.D.; Flatt, P.R.; Hölscher, C.; Gault, V.A. Liraglutide improves hippocampal synaptic plasticity associated with increased expression of Mash1 in ob/ob mice. Int. J. Obes. 2013, 37, 678–684. [Google Scholar] [CrossRef]
  113. Valladolid-Acebes, I.; Merino, B.; Principato, A.; Fole, A.; Barbas, C.; Lorenzo, M.P.; García, A.; Del Olmo, N.; Ruiz-Gayo, M.; Cano, V. High-fat diets induce changes in hippocampal glutamate metabolism and neurotransmission. Am. J. Physiol. Endocrinol. Metab. 2012, 302, E396–E402. [Google Scholar] [CrossRef] [PubMed]
  114. Kuhn, H.G.; Skau, S.; Nyberg, J. A lifetime perspective on risk factors for cognitive decline with a special focus on early events. Cereb. Circ. Cogn. Behav. 2024, 6, 100217. [Google Scholar] [CrossRef] [PubMed]
  115. Mou, Y.; Blok, E.; Barroso, M.; Jansen, P.W.; White, T.; Voortman, T. Dietary patterns, brain morphology and cognitive performance in children: Results from a prospective population-based study. Eur. J. Epidemiol. 2023, 38, 669–687. [Google Scholar] [CrossRef] [PubMed]
  116. Cao, G.Y.; Li, M.; Han, L.; Tayie, F.; Yao, S.S.; Huang, Z.; Ai, P.; Liu, Y.Z.; Hu, Y.H.; Xu, B. Dietary Fat Intake and Cognitive Function among Older Populations: A Systematic Review and Meta-Analysis. J. Prev. Alzheimer’s Dis. 2019, 6, 204–211. [Google Scholar] [CrossRef]
  117. Choi, S.H.; Hong, E.S.; Lim, S. Clinical implications of adipocytokines and newly emerging metabolic factors with relation to insulin resistance and cardiovascular health. Front. Endocrinol. 2013, 4, 97. [Google Scholar] [CrossRef]
  118. Bray, G.A.; Kim, K.K.; Wilding, J.P.H.; World Obesity Federation. Obesity: A chronic relapsing progressive disease process. A position statement of the World Obesity Federation. Obes. Rev. 2017, 18, 715–723. [Google Scholar] [CrossRef]
  119. Kawai, T.; Autieri, M.V.; Scalia, R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am. J. Physiol. Cell. Physiol. 2021, 320, C375–C391. [Google Scholar] [CrossRef]
  120. Kojta, I.; Chacińska, M.; Błachnio-Zabielska, A. Obesity, Bioactive Lipids, and Adipose Tissue Inflammation in Insulin Resistance. Nutrients 2020, 12, 1305. [Google Scholar] [CrossRef]
  121. Knight, Z.A.; Hannan, K.S.; Greenberg, M.L.; Friedman, J.M. Hyperleptinemia is required for the development of leptin resistance. PLoS ONE 2010, 5, e11376. [Google Scholar] [CrossRef] [PubMed]
  122. Mori, H.; Hanada, R.; Hanada, T.; Aki, D.; Mashima, R.; Nishinakamura, H.; Torisu, T.; Chien, K.R.; Yasukawa, H.; Yoshimura, A. Socs3 deficiency in the brain elevates leptin sensitivity and confers resistance to diet-induced obesity. Nat. Med. 2004, 10, 739–743. [Google Scholar] [CrossRef] [PubMed]
  123. Bence, K.K.; Delibegovic, M.; Xue, B.; Gorgun, C.Z.; Hotamisligil, G.S.; Neel, B.G.; Kahn, B.B. Neuronal PTP1B regulates body weight, adiposity and leptin action. Nat. Med. 2006, 12, 917–924. [Google Scholar] [CrossRef] [PubMed]
  124. Wauman, J.; Tavernier, J. Leptin receptor signaling: Pathways to leptin resistance. Front. Biosci. 2011, 16, 2771–2793. [Google Scholar] [CrossRef]
  125. Barnard, N.D.; Bunner, A.E.; Agarwal, U. Saturated and trans fats and dementia: A systematic review. Neurobiol. Aging 2014, 35 (Suppl. S2), S65–S73. [Google Scholar] [CrossRef]
  126. Corley, J.; Cox, S.R.; Taylor, A.M.; Hernandez, M.V.; Maniega, S.M.; Ballerini, L.; Wiseman, S.; Meijboom, R.; Backhouse, E.V.; Bastin, M.E.; et al. Dietary patterns, cognitive function, and structural neuroimaging measures of brain aging. Exp. Gerontol. 2020, 142, 111117. [Google Scholar] [CrossRef]
  127. Sharma, A.N.; Elased, K.M.; Garrett, T.L.; Lucot, J.B. Neurobehavioral deficits in db/db diabetic mice. Physiol. Behav. 2010, 101, 381–388. [Google Scholar] [CrossRef]
  128. Morales, L.; Del Olmo, N.; Valladolid-Acebes, I.; Fole, A.; Cano, V.; Merino, B.; Stucchi, P.; Ruggieri, D.; López, L.; Alguacil, L.F.; et al. Shift of circadian feeding pattern by high-fat diets is coincident with reward deficits in obese mice. PLoS ONE 2012, 7, e36139. [Google Scholar] [CrossRef]
  129. Davidson, T.L.; Chan, K.; Jarrard, L.E.; Kanoski, S.E.; Clegg, D.J.; Benoit, S.C. Contributions of the hippocampus and medial prefrontal cortex to energy and body weight regulation. Hippocampus 2009, 19, 235–252. [Google Scholar] [CrossRef]
  130. Stranahan, A.M.; Mattson, M.P. Impact of energy intake and expenditure on neuronal plasticity. Neuromol. Med. 2008, 10, 209–218. [Google Scholar] [CrossRef]
  131. Eichen, D.M.; Kang Sim, D.E.; Appleton-Knapp, S.L.; Strong, D.R.; Boutelle, K.N. Adults with overweight or obesity use less efficient memory strategies compared to adults with healthy weight on a verbal list learning task modified with food words. Appetite 2023, 181, 106402. [Google Scholar] [CrossRef] [PubMed]
  132. Hayes, A.M.R.; Lauer, L.T.; Kao, A.E.; Sun, S.; Klug, M.E.; Tsan, L.; Rea, J.J.; Subramanian, K.S.; Gu, C.; Tanios, N.; et al. Western diet consumption impairs memory function via dysregulated hippocampus acetylcholine signaling. Brain Behav. Immun. 2024, 118, 408–422. [Google Scholar] [CrossRef] [PubMed]
  133. Avena, N.M.; Bocarsly, M.E.; Hoebel, B.G. Animal models of sugar and fat bingeing: Relationship to food addiction and increased body weight. Methods Mol. Biol. 2012, 829, 351–365. [Google Scholar] [PubMed]
  134. Epstein, L.H.; Biondolillo, M.J.; Rizwan, A.; Ghanim, H.; Dandona, P.; Bickel, W.K.; Paluch, R.A. Insulin Resistance and Glycated Hemoglobin in Obesity Are Associated With Preference for Sugar-Sweetened Yogurt: A Pilot Study. Psychosom. Med. 2023, 85, 289–293. [Google Scholar] [CrossRef]
  135. Greenwood, C.E.; Winocur, G. High-fat diets, insulin resistance and declining cognitive function. Neurobiol. Aging 2005, 26 (Suppl. S1), 42–45. [Google Scholar] [CrossRef]
  136. Lindqvist, A.; Mohapel, P.; Bouter, B.; Frielingsdorf, H.; Pizzo, D.; Brundin, P.; Erlanson-Albertsson, C. High-fat diet impairs hippocampal neurogenesis in male rats. Eur. J. Neurol. 2006, 13, 1385–1388. [Google Scholar] [CrossRef]
  137. Erichsen, J.M.; Fadel, J.R.; Reagan, L.P. Peripheral versus central insulin and leptin resistance: Role in metabolic disorders, cognition, and neuropsychiatric diseases. Neuropharmacology 2022, 203, 108877. [Google Scholar] [CrossRef]
  138. Mota, B.; Brás, A.R.; Araújo-Andrade, L.; Silva, A.; Pereira, P.A.; Madeira, M.D.; Cardoso, A. High-Caloric Diets in Adolescence Impair Specific GABAergic Subpopulations, Neurogenesis, and Alter Astrocyte Morphology. Int. J. Mol. Sci. 2024, 25, 5524. [Google Scholar] [CrossRef]
  139. Del Olmo, N.; Ruiz-Gayo, M. Influence of High-Fat Diets Consumed During the Juvenile Period on Hippocampal Morphology and Function. Front. Cell. Neurosci. 2018, 12, 439. [Google Scholar] [CrossRef]
  140. Freeman, L.R.; Haley-Zitlin, V.; Rosenberger, D.S.; Granholm, A.C. Damaging effects of a high-fat diet to the brain and cognition: A review of proposed mechanisms. Nutr. Neurosci. 2014, 17, 241–251. [Google Scholar] [CrossRef]
  141. Pantiya, P.; Thonusin, C.; Chunchai, T.; Ongnok, B.; Nawara, W.; Arunsak, B.; Chattipakorn, N.; Chattipakorn, S.C. Higher untrained fitness exerts a neuroprotection in Independence to caloric restriction or exercise in high-fat diet-induced obesity. Exp. Neurol. 2023, 365, 114416. [Google Scholar] [CrossRef] [PubMed]
  142. Paulo, S.L.; Miranda-Lourenço, C.; Belo, R.F.; Rodrigues, R.S.; Fonseca-Gomes, J.; Tanqueiro, S.R.; Geraldes, V.; Rocha, I.; Sebastião, A.M.; Xapelli, S.; et al. High Caloric Diet Induces Memory Impairment and Disrupts Synaptic Plasticity in Aged Rats. Curr. Issues Mol. Biol. 2021, 43, 2305–2319. [Google Scholar] [CrossRef] [PubMed]
  143. Akazawa, C.; Shigemoto, R.; Bessho, Y.; Nakanishi, S.; Mizuno, N. Differential expression of five N-methyl-D-aspartate receptor subunit mRNAs in the cerebellum of developing and adult rats. J. Comp. Neurol. 1994, 347, 150–160. [Google Scholar] [CrossRef] [PubMed]
  144. Monyer, H.; Burnashev, N.; Laurie, D.J.; Sakmann, B.; Seeburg, P.H. Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron 1994, 12, 529–540. [Google Scholar] [CrossRef] [PubMed]
  145. Law, A.J.; Weickert, C.S.; Webster, M.J.; Herman, M.M.; Kleinman, J.E.; Harrison, P.J. Changes in NMDA receptor subunit mRNAs and cyclophilin mRNA during development of the human hippocampus. Ann. N. Y. Acad. Sci. 2003, 1003, 426–430. [Google Scholar] [CrossRef]
  146. Huang, X.T.; Yue, S.J.; Li, C.; Guo, J.; Huang, Y.H.; Han, J.Z.; Feng, D.D.; Luo, Z.Q. Antenatal blockade of N-methyl-D-aspartate receptors by Memantine reduces the susceptibility to diabetes induced by a high-fat diet in rats with intrauterine growth restriction. Biol. Reprod. 2017, 96, 960–970. [Google Scholar] [CrossRef] [PubMed]
  147. Hermanussen, M.; Tresguerres, J.A. A new anti-obesity drug treatment: First clinical evidence that, antagonising glutamate-gated Ca2+ ion channels with memantine normalises binge-eating disorders. Econ. Hum. Biol. 2005, 3, 329–337. [Google Scholar] [CrossRef]
  148. Labban, R.S.M.; Alfawaz, H.; Almnaizel, A.T.; Hassan, W.M.; Bhat, R.S.; Moubayed, N.M.; Bjørklund, G.; El-Ansary, A. High-fat diet-induced obesity and impairment of brain neurotransmitter pool. Transl. Neurosci. 2020, 11, 147–160. [Google Scholar] [CrossRef]
  149. Boswell, R.G.; Potenza, M.N.; Grilo, C.M. The Neurobiology of Binge-eating Disorder Compared with Obesity: Implications for Differential Therapeutics. Clin. Ther. 2021, 43, 50–69. [Google Scholar] [CrossRef]
  150. Gómez-Apo, E.; Mondragón-Maya, A.; Ferrari-Díaz, M.; Silva-Pereyra, J. Structural Brain Changes Associated with Overweight and Obesity. J. Obes. 2021, 2021, 6613385. [Google Scholar] [CrossRef]
  151. McCabe, D.P.; Roediger, H.L.; McDaniel, M.A.; Balota, D.A.; Hambrick, D.Z. The relationship between working memory capacity and executive functioning: Evidence for a common executive attention construct. Neuropsychology 2010, 24, 222–243. [Google Scholar] [CrossRef] [PubMed]
  152. Higgs, S.; Spetter, M.S.; Thomas, J.M.; Rotshtein, P.; Lee, M.; Hallschmid, M.; Dourish, C.T. Interactions between metabolic, reward and cognitive processes in appetite control: Implications for novel weight management therapies. J. Psychopharmacol. 2017, 31, 1460–1474. [Google Scholar] [CrossRef] [PubMed]
  153. Davidson, T.L.; Jones, S.; Roy, M.; Stevenson, R.J. The Cognitive Control of Eating and Body Weight: It’s More Than What You “Think”. Front. Psychol. 2019, 10, 62. [Google Scholar] [CrossRef] [PubMed]
  154. Favieri, F.; Forte, G.; Casagrande, M. The Executive Functions in Overweight and Obesity: A Systematic Review of Neuropsychological Cross-Sectional and Longitudinal Studies. Front. Psychol. 2019, 10, 2126. [Google Scholar] [CrossRef] [PubMed]
  155. Yeomans, M.R.; Armitage, R.; Atkinson, R.; Francis, H.; Stevenson, R.J. Habitual intake of fat and sugar is associated with poorer memory and greater impulsivity in humans. PLoS ONE 2023, 18, e0290308. [Google Scholar] [CrossRef]
  156. Flores-Cordero, J.A.; Pérez-Pérez, A.; Jiménez-Cortegana, C.; Alba, G.; Flores-Barragán, A.; Sánchez-Margalet, V. Obesity as a Risk Factor for Dementia and Alzheimer’s Disease: The Role of Leptin. Int. J. Mol. Sci. 2022, 23, 5202. [Google Scholar] [CrossRef]
  157. Patel, V.; Edison, P. Cardiometabolic risk factors and neurodegeneration: A review of the mechanisms underlying diabetes, obesity and hypertension in Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 2024, 95, 581–589. [Google Scholar] [CrossRef]
  158. Maioli, S.; Lodeiro, M.; Merino-Serrais, P.; Falahati, F.; Khan, W.; Puerta, E.; Codita, A.; Rimondini, R.; Ramirez, M.J.; Simmons, A.; et al. Alterations in brain leptin signalling in spite of unchanged CSF leptin levels in Alzheimer’s disease. Aging Cell 2015, 14, 122–129. [Google Scholar] [CrossRef]
  159. Elinav, E.; Niv-Spector, L.; Katz, M.; Price, T.O.; Ali, M.; Yacobovitz, M.; Solomon, G.; Reicher, S.; Lynch, J.L.; Halpern, Z.; et al. Pegylated leptin antagonist is a potent orexigenic agent: Preparation and mechanism of activity. Endocrinology 2009, 150, 3083–3091. [Google Scholar] [CrossRef]
  160. Malekizadeh, Y.; Holiday, A.; Redfearn, D.; Ainge, J.A.; Doherty, G.; Harvey, J. A Leptin Fragment Mirrors the Cognitive Enhancing and Neuroprotective Actions of Leptin. Cereb. Cortex 2017, 27, 4769–4782. [Google Scholar] [CrossRef]
  161. Doherty, G.; Holiday, A.; Malekizadeh, Y.; Manolescu, C.; Duncan, S.; Flewitt, I.; Hamilton, K.; MacLeod, B.; Ainge, J.A.; Harvey, J. Leptin-based hexamers facilitate memory and prevent amyloid-driven AMPA receptor internalisation and neuronal degeneration. J. Neurochem. 2023, 165, 809–826. [Google Scholar] [CrossRef] [PubMed]
  162. Matochik, J.A.; London, E.D.; Yildiz, B.O.; Ozata, M.; Caglayan, S.; DePaoli, A.M.; Wong, M.L.; Licinio, J. Effect of leptin replacement on brain structure in genetically leptin-deficient adults. J. Clin. Endocrinol. Metab. 2005, 90, 2851–2854. [Google Scholar] [CrossRef]
  163. Dalamaga, M.; Chou, S.H.; Shields, K.; Papageorgiou, P.; Polyzos, S.A.; Mantzoros, C.S. Leptin at the intersection of neuroendocrinology and metabolism: Current evidence and therapeutic perspectives. Cell Metab. 2013, 18, 29–42. [Google Scholar] [CrossRef] [PubMed]
  164. Paz-Filho, G.J. The Effects of Leptin Replacement on Neural Plasticity. Neural Plast. 2016, 2016, 8528934. [Google Scholar] [CrossRef] [PubMed]
  165. Lu, X.Y.; Kim, C.S.; Frazer, A.; Zhang, W. Leptin: A potential novel antidepressant. Proc. Natl. Acad. Sci. USA 2006, 103, 1593–1598. [Google Scholar] [CrossRef]
  166. London, E.D.; Berman, S.M.; Chakrapani, S.; Delibasi, T.; Monterosso, J.; Erol, H.K.; Paz-Filho, G.; Wong, M.L.; Licinio, J. Short-term plasticity of gray matter associated with leptin deficiency and replacement. J. Clin. Endocrinol. Metab. 2011, 96, E1212–E1220, Erratum in J. Clin. Endocrinol. Metab. 2011, 96, 3576. [Google Scholar] [CrossRef]
  167. Garza, J.C.; Guo, M.; Zhang, W.; Lu, X.Y. Leptin increases adult hippocampal neurogenesis in vivo and in vitro. J. Biol. Chem. 2008, 283, 18238–18247. [Google Scholar] [CrossRef]
  168. Irving, A.J.; Harvey, J. Leptin regulation of hippocampal synaptic function in health and disease. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2013, 369, 20130155. [Google Scholar] [CrossRef]
  169. Forny-Germano, L.; De Felice, F.G.; Vieira, M.N.D.N. The Role of Leptin and Adiponectin in Obesity-Associated Cognitive Decline and Alzheimer’s Disease. Front. Neurosci. 2019, 12, 1027. [Google Scholar] [CrossRef]
  170. DeFronzo, R.A.; Ferrannini, E.; Groop, L.; Henry, R.R.; Herman, W.H.; Holst, J.J.; Hu, F.B.; Kahn, C.R.; Raz, I.; Shulman, G.I.; et al. Type 2 diabetes mellitus. Nat. Rev. Dis. Primers 2015, 1, 15019. [Google Scholar] [CrossRef]
  171. Müller, T.D.; Blüher, M.; Tschöp, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances and challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef] [PubMed]
  172. Moghazy, H.M.; Abdelhaliem, N.G.; Mohammed, S.A.; Hassan, A.; Abdelrahman, A. Liraglutide versus pramlintide in protecting against cognitive function impairment through affecting PI3K/AKT/GSK-3β/TTBK1 pathway and decreasing Tau hyperphosphorylation in high-fat diet-streptozocin rat model. Pflug. Arch. 2024, 476, 779–795. [Google Scholar] [CrossRef] [PubMed]
  173. Patrick, S.; Corrigan, R.; Grizzanti, J.; Mey, M.; Blair, J.; Pallas, M.; Camins, A.; Lee, H.G.; Casadesus, G. Neuroprotective Effects of the Amylin Analog, Pramlintide, on Alzheimer’s Disease Are Associated with Oxidative Stress Regulation Mechanisms. J. Alzheimer’s Dis. 2019, 69, 157–168. [Google Scholar] [CrossRef]
  174. Qiu, W.Q.; Zhu, H. Amylin and its analogs: A friend or foe for the treatment of Alzheimer’s disease? Front. Aging Neurosci. 2014, 6, 186. [Google Scholar] [CrossRef]
  175. D’Ascanio, A.M.; Mullally, J.A.; Frishman, W.H. Cagrilintide: A Long-Acting Amylin Analog for the Treatment of Obesity. Cardiol. Rev. 2024, 32, 83–90. [Google Scholar] [CrossRef] [PubMed]
  176. Enebo, L.B.; Berthelsen, K.K.; Kankam, M.; Lund, M.T.; Rubino, D.M.; Satylganova, A.; Lau, D.C.W. Safety, tolerability, pharmacokinetics, and pharmacodynamics of concomitant administration of multiple doses of cagrilintide with semaglutide 2.4 mg for weight management: A randomised, controlled, phase 1b trial. Lancet 2021, 397, 1736–1748. [Google Scholar] [CrossRef]
  177. Gadde, K.M.; Allison, D.B. Long-acting amylin analogue for weight reduction. Lancet 2021, 398, 2132–2134. [Google Scholar] [CrossRef]
  178. Lau, D.C.W.; Erichsen, L.; Francisco, A.M.; Satylganova, A.; le Roux, C.W.; McGowan, B.; Pedersen, S.D.; Pietiläinen, K.H.; Rubino, D.; Batterham, R.L. Once-weekly cagrilintide for weight management in people with overweight and obesity: A multicentre, randomised, double-blind, placebo-controlled and active-controlled, dose-finding phase 2 trial. Lancet 2021, 398, 2160–2172. [Google Scholar] [CrossRef]
  179. Becerril, S.; Frühbeck, G. Cagrilintide plus semaglutide for obesity management. Lancet 2021, 397, 1687–1689. [Google Scholar] [CrossRef]
  180. Wu, T.; Gao, X.; Chen, M.; van Dam, R.M. Long-term effectiveness of diet-plus-exercise interventions vs. diet-only interventions for weight loss: A meta-analysis. Obes. Rev. 2009, 10, 313–323. [Google Scholar] [CrossRef]
  181. Miller, C.T.; Fraser, S.F.; Levinger, I.; Straznicky, N.E.; Dixon, J.B.; Reynolds, J.; Selig, S.E. The effects of exercise training in addition to energy restriction on functional capacities and body composition in obese adults during weight loss: A systematic review. PLoS ONE 2013, 8, e81692. [Google Scholar] [CrossRef]
  182. Khalafi, M.; Azali Alamdari, K.; Symonds, M.E.; Rohani, H.; Sakhaei, M.H. A comparison of the impact of exercise training with dietary intervention versus dietary intervention alone on insulin resistance and glucose regulation in individual with overweight or obesity: A systemic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2023, 63, 9349–9363. [Google Scholar] [CrossRef] [PubMed]
  183. Lecoultre, V.; Ravussin, E.; Redman, L.M. The fall in leptin concentration is a major determinant of the metabolic adaptation induced by caloric restriction independently of the changes in leptin circadian rhythms. J. Clin. Endocrinol. Metab. 2011, 96, E1512–E1516. [Google Scholar] [CrossRef] [PubMed]
  184. Rodrigues, K.C.D.C.; Pereira, R.M.; de Campos, T.D.P.; de Moura, R.F.; da Silva, A.S.R.; Cintra, D.E.; Ropelle, E.R.; Pauli, J.R.; de Araújo, M.B.; de Moura, L.P. The Role of Physical Exercise to Improve the Browning of White Adipose Tissue via POMC Neurons. Front. Cell. Neurosci. 2018, 12, 88. [Google Scholar] [CrossRef] [PubMed]
  185. Li, C.; Meng, F.; Lei, Y.; Liu, J.; Liu, J.; Zhang, J.; Liu, F.; Liu, C.; Guo, M.; Lu, X.Y. Leptin regulates exon-specific transcription of the Bdnf gene via epigenetic modifications mediated by an AKT/p300 HAT cascade. Mol. Psychiatry 2021, 26, 3701–3722. [Google Scholar] [CrossRef]
  186. Yu, W.H.; Kimura, M.; Walczewska, A.; Karanth, S.; McCann, S.M. Role of leptin in hypothalamic-pituitary function. Proc. Natl. Acad. Sci. USA 1997, 94, 1023–1028. [Google Scholar] [CrossRef] [PubMed]
  187. Ahima, R.S.; Prabakaran, D.; Mantzoros, C.; Qu, D.; Lowell, B.; Maratos-Flier, E.; Flier, J.S. Role of leptin in the neuroendocrine response to fasting. Nature 1996, 382, 250–252. [Google Scholar] [CrossRef]
  188. Harvey, J. Novel Leptin-Based Therapeutic Strategies to Limit Synaptic Dysfunction in Alzheimer’s Disease. Int. J. Mol. Sci. 2024, 25, 7352. [Google Scholar] [CrossRef]
  189. Horie, N.C.; Serrao, V.T.; Simon, S.S.; Gascon, M.R.; Dos Santos, A.X.; Zambone, M.A.; Del Bigio de Freitas, M.M.; Cunha-Neto, E.; Marques, E.L.; Halpern, A.; et al. Cognitive Effects of Intentional Weight Loss in Elderly Obese Individuals With Mild Cognitive Impairment. J. Clin. Endocrinol. Metab. 2016, 101, 1104–1112. [Google Scholar] [CrossRef]
  190. Wittekind, D.A.; Kratzsch, J.; Mergl, R.; Baber, R.; Wirkner, K.; Schroeter, M.L.; Witte, A.V.; Villringer, A.; Kluge, M. Leptin, but not ghrelin, is associated with food addiction scores in a population-based subject sample. Front. Psychiatry 2023, 14, 1200021. [Google Scholar] [CrossRef]
  191. Peters, T.; Antel, J.; Föcker, M.; Esber, S.; Hinney, A.; Schéle, E.; Dickson, S.L.; Albayrak, Ö.; Hebebrand, J. The association of serum leptin levels with food addiction is moderated by weight status in adolescent psychiatric inpatients. Eur. Eat. Disord. Rev. 2018, 26, 618–628. [Google Scholar] [CrossRef] [PubMed]
  192. World Health Organization (WHO). Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 14 October 2024).
Figure 1. Synoptic overview of the hippocampal regulatory actions of leptin in both health and disease. In a healthy state (left), leptin is secreted from adipocytes at a physiological level and easily crosses the blood–brain barrier through an ObRa-dependent saturable transport process to exert its effects on the brain [24,28]. Within the hippocampus, leptin promotes cognitive function by swiftly modulating neuronal survival and synaptic plasticity. However, in obesity (right), leptin levels are elevated, resulting in leptin resistance and diminished transport into the brain. This impaired leptin signaling in the hippocampus is linked to disrupted neuronal function and synaptic plasticity, thus increasing the risk of cognitive decline. Created with Biorender.com.
Figure 1. Synoptic overview of the hippocampal regulatory actions of leptin in both health and disease. In a healthy state (left), leptin is secreted from adipocytes at a physiological level and easily crosses the blood–brain barrier through an ObRa-dependent saturable transport process to exert its effects on the brain [24,28]. Within the hippocampus, leptin promotes cognitive function by swiftly modulating neuronal survival and synaptic plasticity. However, in obesity (right), leptin levels are elevated, resulting in leptin resistance and diminished transport into the brain. This impaired leptin signaling in the hippocampus is linked to disrupted neuronal function and synaptic plasticity, thus increasing the risk of cognitive decline. Created with Biorender.com.
Biomedicines 12 02422 g001
Figure 2. Physiological leptin signal transduction and its effects in the hypothalamus and hippocampus. Upon leptin binding to ObRb (middle), Jak2 undergoes phosphorylation, triggering the dimerization and nuclear translocation of STAT3 (1). Concurrently, the PI3K/Akt (2) and MAPK (3) pathways are activated following Jak2 phosphorylation, resulting in nuclear gene transcription. ObRb signaling is negatively regulated by the activation of SOCS3 and PTP1B (4). Leptin receptor signaling in the hypothalamus (left) stimulates the anorexigenic POMC/CART neurons. The release and further cleavage of POMC yields the melanocortin-stimulating hormone (α-MSH), which signals through melanocortin receptors, leading to anorexigenic effects. In contrast, leptin signal transduction represses the expression of the orexigenic neuropeptides NPY/AgRP in neighboring neurons. AgRp also acts as an inverse agonist of melanocortin receptors, counteracting the effects of α-MSH. To regulate energy homeostasis, these first-order neuronal targets of adiposity signals project from the arcuate nucleus (ARC) to the paraventricular nucleus (PVN) and other regions of the brain. As a consequence, food intake is reduced in favor of increased energy expenditure. In the hippocampus (right), activation of hippocampal ObRb enhances NMDA receptor (NMDA-R) function, leading to modifications in AMPA receptor (AMPA-R) trafficking, which subsequently promotes sustained changes in excitatory synaptic strength. Abbreviations: ERK1/2, extracellular signal-regulated kinase 1/2; ER, endoplasmic reticulum; Grb2, growth factor receptor-bound protein 2; MCR-4, melanocortin receptor 4; MEK, mitogen-activated protein kinase; Raf, proto-oncogene serine/threonine-protein kinase; Ras, rat sarcoma virus small GTPase; SHP-2, Src homology region 2 domain-containing phosphatase-2; SOS, Son of Sevenless guanine nucleotide exchange factor. Created with Biorender.com.
Figure 2. Physiological leptin signal transduction and its effects in the hypothalamus and hippocampus. Upon leptin binding to ObRb (middle), Jak2 undergoes phosphorylation, triggering the dimerization and nuclear translocation of STAT3 (1). Concurrently, the PI3K/Akt (2) and MAPK (3) pathways are activated following Jak2 phosphorylation, resulting in nuclear gene transcription. ObRb signaling is negatively regulated by the activation of SOCS3 and PTP1B (4). Leptin receptor signaling in the hypothalamus (left) stimulates the anorexigenic POMC/CART neurons. The release and further cleavage of POMC yields the melanocortin-stimulating hormone (α-MSH), which signals through melanocortin receptors, leading to anorexigenic effects. In contrast, leptin signal transduction represses the expression of the orexigenic neuropeptides NPY/AgRP in neighboring neurons. AgRp also acts as an inverse agonist of melanocortin receptors, counteracting the effects of α-MSH. To regulate energy homeostasis, these first-order neuronal targets of adiposity signals project from the arcuate nucleus (ARC) to the paraventricular nucleus (PVN) and other regions of the brain. As a consequence, food intake is reduced in favor of increased energy expenditure. In the hippocampus (right), activation of hippocampal ObRb enhances NMDA receptor (NMDA-R) function, leading to modifications in AMPA receptor (AMPA-R) trafficking, which subsequently promotes sustained changes in excitatory synaptic strength. Abbreviations: ERK1/2, extracellular signal-regulated kinase 1/2; ER, endoplasmic reticulum; Grb2, growth factor receptor-bound protein 2; MCR-4, melanocortin receptor 4; MEK, mitogen-activated protein kinase; Raf, proto-oncogene serine/threonine-protein kinase; Ras, rat sarcoma virus small GTPase; SHP-2, Src homology region 2 domain-containing phosphatase-2; SOS, Son of Sevenless guanine nucleotide exchange factor. Created with Biorender.com.
Biomedicines 12 02422 g002
Table 1. Ongoing clinical trials in obesity and/or overweight using amylin analogs or Celastrol (up to 16 October 2024).
Table 1. Ongoing clinical trials in obesity and/or overweight using amylin analogs or Celastrol (up to 16 October 2024).
NCT Number
Phase/Design
Drug Name(s)Condition(s)Primary Outcome(s)SponsorStatusLast Updated Posted
Pramlintide
1NCT03560960
Early Phase I/Diagnostic
Pramlintide (Symlin) (monotherapy) Alzheimer´s Disease; Mild Cognitive Impairment; Plasma Aβ and t-tau changes.
Plasma inflammatory changes.
Metabolic changes in blood.
All parameters determined 5, 30, 60, and 180 min after a Pramlintide challenge test.
Boston UniversityRecruiting31 July 2024
2NCT00691158
N/A/RTBPC
Pramlintide (monotherapy or combined with Metreleptin) or placeboObesityHypothalamic, brainstem and whole brain fMRI response measurements to different treatments.Oregon Health and Science UniversityActive, not recruiting16 December 2022
3NCT00690235
IV RDBPC
Pramlintide (monotherapy) or placebo Schizophrenia; Schizoaffective Disorder; Diabetes; Weight GainWeight loss from Pramlintide in Olanzapine- or Clozapine-induced weight gain patients with schizophreniaUniversity of Texas Southwestern Medical CenterCompleted16 November 2018
4NCT00189514
II/RDBPC
Pramlintide acetate (monotherapy) or placebo ObesityLong-term effects of Pramlintide on body weight in obese subjects.
Long-term safety and tolerability of the drug in obese patients.
AstraZenecaCompleted11 June 2015
5NCT00444561
II/RDBPC
Pramlintide acetate (AC137, monotherapy) or placeboOverweight; ObesityPharmacodynamics of Pramlintide in obese subjects.AstraZenecaCompleted11 June 2015
6NCT00673387
II/RDBPC
Pramlintide acetate (Smylin) or placebo versus Metreleptin or placeboOverweight; ObesityMeasurement of the mean percentage of change in body weight from baseline to week 28 of the study.AstraZenecaCompleted15 April 2015
7NCT00819234
II/RTBPC
Pramlintide (combined with Metreleptin) or placeboObesityMeasurement of the mean percentage of change in body weight from the baseline of the original study (NCT00673387) at week 52 in the extension study.AstraZenecaCompleted15 April 2015
8NCT01235741
II/RDBPC
Pramlintide (combined with Metreleptin) or placeboObesityEffect of 16 weeks of treatment on body weight in subjects with obesity.
Studies of safety and tolerability of the drug combination.
AstraZenecaTerminated15 April 2015
9NCT0039292
IIa/RDBPC
Pramlintide acetate (Smylin) or placebo; Metreleptin or placeboOverweight; ObesityEffect of 16 weeks of treatment on body weight in participants with overweight and/or obesity.AstraZenecaCompleted14 April 2015
10NCT00112021
IIb/RDBPC
Pramlintide acetate (monotherapy) or placebo ObesityEffect of 16 weeks of treatment on body weight in subjects with obesity.
Studies of safety and tolerability of Pramlintide in participants with obesity.
AstraZenecaCompleted10 April 2015
11NCT00402077
II/RSBPC
Pramlintide acetate (Symlin) compared to Sibutramine, Phentermine or placeboOverweight; ObesityDetermination of treatment-related adverse events during 24-week treatment period.
Change in body weight from baseline to week 12 of treatment.
AstraZenecaCompleted6 March 2015
Cagrilintide
12NCT06131372
II/RQBPC
CagriSema or placebo versus active comparators [Cagrilintide or Semaglutide, monotherapies] or placeboChronic Kidney Disease; Type 2 Diabetes; Obesity Change in urinary albumin-to-creatinine ratio (UACR) from baseline (week 0) to end of treatment (week 26).Novo Nordisk A/SRecruiting26 August 2024
13NCT06388187
III/RQBPC
CagriSema (two different doses) or placebo (two diferent doses)Obesity Body weight changes from baseline (week 0) to end of treatment (week 68).
Achievement of ≥5% weight reduction from baseline to the end of the treatmen.
Novo Nordisk A/SRecruiting20 August 2024
14NCT06131437
III/ROL
CagriSema versus active comparator (Tirzepatide) Obesity Change in body weight from baseline (week 0) to end of treatment (week 72).Novo Nordisk A/SActive, not recruiting20 August 2024
15NCT05567796 (REDEFINE 1)
III/RQBPC
CagriSema or placebo versus active comparators [Cagrilintide or Semaglutide, monotherapies] ObesityBody weight changes from baseline (week 0) to end of treatment (week 68).
Achievement of ≥5% weight reduction from baseline to the end of the treatment.
Novo Nordisk A/SActive, not recruiting20 August 2024
16NCT05813925
III/RQBPC
CagriSema versus active comparator (Semaglutide)Overweight; ObesityBody weight changes from baseline (week 0) to end of treatment (week 68).Novo Nordisk A/SRecruiting6 August 2024
17NCT05394519 (REDEFINE 2)
III/RQBPC
CagriSema or placeboOverweight; Obesity; Type 2 Diabetes Mellitus Body weight changes from baseline (week 0) to end of treatment (week 68).
Achievement of ≥5% weight reduction from baseline to the end of the treatment.
Novo Nordisk A/SActive, not recruiting17 July 2024
18NCT05996848 (REDEFINE 6)
III/RDBPC
CagriSema versus active comparator [Semaglutide, monotherapy] or placeboOverweight; ObesityBody weight changes from baseline (week 0) to end of treatment (week 68).
Achievement of ≥5% weight reduction from baseline to the end of the treatment.
Novo Nordisk A/SRecruiting1 July 2024
19NCT05804162
I/RQBPC
Cagrilintide or placebo versus Moxifloxacin and placeboObesityChange from baseline in Fredericia’s heart rate-corrected QT interval (QTcF).Novo Nordisk A/SCompleted30 October 2023
20NCT04940078
I/ROL
Cagrilintide (monotherapy) versus CagriSema (Cagrilintide+Semaglutide)Overweight; ObesityPharmacokinetic characterization of treatments in the subject population.Novo Nordisk A/SCompleted20 February 2024
21NCT06289504
I/ROL
CagriSema +Atorvastatin + Warfarin versus Semaglutide +Atorvastatin + WarfarinObesityPharmacokinetic characterization of treatments in the subject population.Novo Nordisk A/SRecruiting14 May 2024
22NCT06207877
I/RQBPC
CagriSema or placeboObesity Changes in energy intake during ad libitum lunch, evening meal and snackbox from baseline to day 156 of the study.Novo Nordisk A/SRecruiting15 March 2024
23NCT06267092
I/RQBPC
CagriSema or placeboOverweight; ObesityChange in mean postprandial appetite score based on visual analog scale (VAS) from baseline to week 24 of the study.Novo Nordisk A/SRecruiting28 February 2024
Davalintide (AC2307)
24NCT00785408
II/RDBPC
AC2307 or placeboOverweight; ObesityChanges in body weight from baseline to week 24 of the study.
Studies of safety and tolerability.
AstraZenecaCompleted19 January 2015
GUBamy (GUB014295)
25NCT06144684
I/RDBPC
GUB014295 (GUBamy) or placeboHealthy Volunteers; OverweightAdverse events (AE) incidence.
Changes in vital signs.
Safety laboratory parameters.
All safety parameters determined from baseline (day 0) to the end of the trial’s part 1 (day 29), part 2A (day 64) and part 2B (day 106).
Gubra A/SRecruiting21 August 2024
Celastrol
26NCT05494112
N/A/Safety OL
Celastrol (dietary supplement)Healthy VolunteersEffect of Celastrol in on liver function.Legend Labz, Inc.Unknown status9 September 2022
Abbreviations: OL, open label; RDPBC, randomized double-blind placebo-controlled; ROL, randomized open-label; RQPBC, randomized quadruple-blind placebo-controlled; RTPBC, randomized triple-blind placebo-controlled.
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

Valladolid-Acebes, I. Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines 2024, 12, 2422. https://doi.org/10.3390/biomedicines12112422

AMA Style

Valladolid-Acebes I. Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines. 2024; 12(11):2422. https://doi.org/10.3390/biomedicines12112422

Chicago/Turabian Style

Valladolid-Acebes, Ismael. 2024. "Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications" Biomedicines 12, no. 11: 2422. https://doi.org/10.3390/biomedicines12112422

APA Style

Valladolid-Acebes, I. (2024). Hippocampal Leptin Resistance and Cognitive Decline: Mechanisms, Therapeutic Strategies and Clinical Implications. Biomedicines, 12(11), 2422. https://doi.org/10.3390/biomedicines12112422

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

Article Metrics

Back to TopTop