Open access peer-reviewed chapter

Biodiversity of the Adipocyte-Derived Hormone, Leptin

Written By

Reji Manjunathan, Dharanibalan Kasiviswanathan and Selvaraj Jayaraman

Submitted: 12 September 2021 Reviewed: 21 September 2021 Published: 09 October 2021

DOI: 10.5772/intechopen.100576

From the Edited Volume

Weight Management - Challenges and Opportunities

Edited by Hassan M. Heshmati

Chapter metrics overview

306 Chapter Downloads

View Full Metrics

Abstract

The adipocyte derived hormone leptin is known for its pivotal role in the regulation of a variety of physiological functions mainly associated with metabolism and energy homeostasis. One of the major functions of leptin is pertain with its angiogenic induction in support of organ development as well as under pathological conditions such as atherosclerosis and cancer. Leptin is a well-known pro-angiogenic growth factor which exerts its role through Ob-R receptor present on endothelial cells. The therapeutic application of leptin is based on its potential to maintain various functions at pathological conditions. In this book chapter, the multi-diversity potentials of leptin are discussed in detail.

Keywords

  • Leptin
  • obesity
  • angiogenesis
  • tumor progression
  • multi-signaling pathways

1. Introduction

Leptin is a 16 kDa non-glycosylated protein derived from adipose tissue, primarily by adipocytes. Leptin is a well-known mediator for food intake and weight loss [1]. Leptin mediates its functions mainly through the receptor located in the hypothalamus and activates signaling cascades associated with energy intake [2]. It circulates through the bloodstream, engages with normal metabolism, regulates energetic homeostasis, reproductive system, and influences the circadian cycle, lipid inflammation, and carbohydrate mechanisms. Leptin is well known for its pro-angiogenic potential and operates multiple signaling agencies through the receptor located in endothelial cells [3, 4]. Leptin is also secrets by other organs, such as the placenta, bone marrow, ovaries, stomach, and cellular structures, including mammary epithelial cells, P/D1 cells, and gastric chief cells [1]. Research has demonstrated that leptin plays a crucial role in maintaining the normal physiology of various vital systems such as the reproductive system and could balance cell proliferation (Figure 1). The pleiotropic hormone also could repair tissue damage and can prevent on-adipocyte lipotoxicity. Though leptin is highly acceptable for its protective mode of action, increased leptin level is often observed in several inflammatory conditions [5]. Hence, a therapeutic approach based on leptin and receptor has become the need of the hour to balance many inflammatory diseases in the human body. The particular book chapter provides an insight into the multi-diversity properties of the pleiotropic hormone leptin.

Figure 1.

Leptin exerts its pleiotropic impact on various organs. It maintains muscle tone and regulates cardiac function. Leptin regulates food intake and body weight management through binding with receptor located at brain. In the thyroid gland, leptin stimulates the secretion of TSH. In the female reproductive system, leptin manage menstrual cycle and supports in embryo implantation. Leptin inhibits insulin secretion and maintains blood-glucose level. Leptin induces sprouting of new vessels from existing ones and enhances ECs proliferation and migration and also it regulates bone metabolism and density.

Advertisement

2. Leptin synthesis and regulation

Leptin derives from adipose tissue’s obese gene (OB) transcription product [6]. The OB gene function was first identified in the ob/ob obese mice model and is located on chromosome 7 (7q31.3) and has three exons and two introns (18 kb) [7, 8]. Leptin receptors are located on chromosome 1 (1p31) and are noted with 17 introns and 18 exons and encode two proteins of 166 and 1162 amino acids, respectively [9]. Leptin receptors are highly expressed in the hypothalamus, cerebellum, and other tissues associated with the vasculature, stomach, and placental organs [10]. Leptin receptors have five spliced isoforms, the longest form expressed in neuronal tissue and the short forms expressed almost in all tissue types [11]. Leptin receptors (OB-R) are structurally similar to the class I cytokine family receptors. Alternative splicing of leptin receptor RNA results in various isoforms, designated as OB-Ra, OB-Rb, OB-Rc, OB-Rd, OB-Re, and OB-Rf. They all have an extracellular domain of more than 800 amino acids, a transmembrane domain of 34 amino acids, and a variable intracellular domain. The pleiotropic biological effects of leptin are explained based on the wide distribution of leptin receptors in humans [5]. Leptin bind to its hypothalamic receptors (Ob-Rs) in the brain and activates appetite and satiety. The concentration of leptin in plasma depends on the person’s dietary behavior, gender, and physical activities. The other hormonal constituents, such as insulin, estrogen, and glucocorticoids, can also influence the regulation mode and the level of leptin in the blood [12, 13]. On the other hand, low energy or fasting, thyroid hormones, androgens, inflammatory cytokines, and adrenergic agonists can inhibit leptin secretion [14].

Advertisement

3. Leptin signaling pathways

Leptin mediates its biological effects by binding to its various alternatively spliced isoforms receptor located at the brain and peripheral tissues. The binding of leptin to its long-form of receptor activates various intracellular signaling pathways, including insulin receptor substrate (IRS)/phosphatidylinositol 3 kinase (PI3K), Janus kinase 2 (JAK2)/Signal transducer, and activator of transcription 3 (STAT3), SH2-containing protein tyrosine phosphatase 2 (SHP2)/Mitogen-activated protein kinase (MAPK), and 5′ adenosine monophosphate-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) [8]. The binding of leptin to its receptor activates JAK2, which in turn phosphorylates the Tyrosine amino acid residues in LepRb and is terminated by a suppressor of cytokine signaling 3 (SOC3) [15].

Leptin input a significant role in energy homeostasis and neuroendocrine function through JAK2/STAT3 signaling pathway. A selective deletion in LepRb or STAT3 in LepRb-expressing neurons ends with obesity and hyperphagia, which further supports the dominant role of the JAK2/STAT3 signaling pathway in the leptin-induced body weight regulation [16]. One interesting fact about leptin and insulin is that both have similar intracellular signaling pathways (PI3K/Akt) in neurons [17]. The ERK, a member of the MAPK family, acts downstream of LepRb and is mediated through SHP2 or by JAK2. Inhibition of ERK prevents leptin-based sympathetic function in brown adipose, which further supports SHP2/MAPK in leptin energy expenditure and food intake [18]. Leptin’s suppressive mode of action on food intake initiates by inhibiting the effect of AMPK in the brain. The inhibition of AMPK regulates feeding through the mTOR (mammalian target of rapamycin)/s6Kinase pathway [19]. In skeletal muscle, leptin directly exists its effect through AMPK signaling and stimulates fatty acid oxidation and glucose uptake [20]. Leptin has a prominent role in the modulation of both innate and adaptive immunity. It stimulates neutrophil chemotaxis and promotes phagocytosis of macrophages through the receptor binding mechanism. It is also known to increase the production of IL-6 and TNF-alpha under pathological conditions [21]. Leptin protective action on neutrophils exerts through PI3K and MAPK depending on signaling and prevents apoptosis of neutrophils. Leptin via STAT3 activation promotes natural killer cell activation [22]. In the adaptive immune response, leptin promotes native T cells proliferation by increasing the expression of interferon-gamma and TNF-alpha in T cells [23].

Apart from the mentioned direct signaling pathways, leptin interacts with many signaling functions as a multifunctional cytokine. Leptin shows a potential functional relationship with Nitric Oxide (NO) and favors NO-mediated lipolysis and vascular tone [24]. The significant other functions of leptin are associated with its predominant role in angiogenesis. It is observed that Endothelial cells (ECs) express OB-R leptin receptors and the binding of leptin to OB-R enables the growth of small blood vessels [3]. Recently, it has been identified that leptin could induce PI3K/Akt/mTOR/s6Kinase signaling pathway and enhance VEGF mRNA’s transcription level while inducing angiogenesis [4]. One of the intriguing possibilities of leptin is that it promotes neovascularization through paracrine mode concerning the volume of fat mass [25]. Leptin could promote proliferation in colonic epithelial cells in vitro conditions. Moreover, the presence of OB-R receptor in human colon cancer cell lines and human Colonia tissue thus supports the angiogenic role of leptin under cancer environment through PI3K/AKT, MAPK/ERK, and JAK2/STAT3 pathways [26, 27]. Leptin could induce apoptosis and regulate actin-myosin cytoskeleton associated with Rho family GTPases (Figure 2) [28].

Figure 2.

Leptin regulates many signaling pathways through receptor (Ob-Rb) binding mechanism. It regulates gene expression through JAK/STAT3 pathway, modulates other signals through PI3K/rho family dependent pathway, induces vasodilation through NO-dependent pathway, and accelerates angiogenesis through PI3K/Akt/mTOR/s6 kinase/VEGF a and PI3/Akt/NO-dependent pathways. Promotes tumor progression through HIF-1 alpha pathway and enhances platelet aggregation through the PLC pathway. The SOS3 molecule function as a regulator of leptin induced signaling activations by negative feedback mechanism.

Advertisement

4. Leptin as an energy balancer

Leptin acts in the brain and maintains energy homeostasis through a negative feedback mechanism [29]. The process is mediated through the receptors in the hypothalamic area named the paraventricular nucleus, ventromedial hypothalamic nucleus, lateral hypothalamic area (LHA), and arcuate nucleus (ARC). The ARC is the primary site for leptin to integrate peripheral energy balance signals [30]. Recently, it has been observed that leptin could play a significant role in the long-term regulation of energy balance and short-term management of body weight and food intake. The gastric leptin produced because of the actions of the intestinal peptide serves as a local stimulus and plays a vital role in food digestion and absorption [31]. The particular area requires more investigations to prove the role of gastric leptin in food digestion and absorption. Research supported the predominant role of leptin in neuroendocrine mediated starvation through changing sympathetic nervous system activity [32].

Overweight or obesity is characterized by increased fat mass and is proportional to circulating leptin levels in individuals [33]. The elevated levels of leptin in body fluid are explained based on leptin resistance. The hypothesis was proved using rodents fed with a high-fat diet and leptin sensitivity loss in ARC neurons [34]. At the cellular level, the inflammatory signals mediate the process of leptin resistance. The two significant characteristics of obesity connect with hyper-leptinemia and leptin resistance. At the molecular level, the leptin gene is over-expressed in overweight or obese individuals [35]. Apart from these functions, many researchers reported the genetic and epigenetic factors that control leptin action in energy homeostasis and food intake [36, 37, 38]. A better understanding of leptin-induced pathogenicity of obesity and obesity-related disorders and the regulation of energy homeostasis will provide an alternative solution in preventing obesity and obesity-related co-morbidities.

Advertisement

5. Leptin as an immune modulator

Despite nutritional regulation, leptin has gained more attention for its pivotal role in inflammation. The innate immune system plays a major role in the regulation of leptin production. Leptin responds to immune cells and its receptors, expressed by most cells, and activates pro-inflammatory features in the host [39]. Leptin plays an essential role in T cell development, and leptin deficiency directly impacts the levels of circulating T cells [40]. Many studies supported the role of leptin in immunity modulation and mentioned the signaling pathways related to the notion [39, 41, 42]. Leptin could accelerate the proliferation process in native CD4+T cells and favored by reducing the levels of IFN from T cells [43, 44]. During the wound healing process, leptin activates both inflammatory and proliferative phases in favor of tissue repair [45]. The increased plasma leptin level acts as an indicator of leptin-induced inflammatory response at the injury site. These exciting features of leptin gained attention as a pro-angiogenic molecule in ischemic tissues [46]. Leptin induces monocyte chemoattractant protein1 (MCP1) expression [47].

Leptin plays a vital role in producing GM-CSF and G-CSF and activating hematopoietic cells in humans [48]. In animal models, up-regulation of leptin has been found in acute inflammation states. But, experimental evidence from rodents does not match with human studies [49]. Leptin plays a significant role in basophils and eosinophils functions and acts as a chemoattractant [50]. Leptin is abnormally expressed in autoimmune diseases, particularly in skin disorders [51]. Obesity decorates skin normal physiology such as keratosis pilaris, tags, and striae diseases and increases the levels of pro-inflammatory cytokines and adipokines, including leptin [52, 53]. In the event of inflammation, leptin increases the release of Nitric Oxide and activates the macrophages and neutrophils, and increases natural killer cells’ activity (NK) [54]. Leptin up-regulates the cytokines production and phagocytic function in obese conditions [55]. It balances monocytes and activations markers and directly involves in interleukin1 and cyclooxygenase expression [56]. One of the prominent roles of leptin pertains to maintaining the balance between the immune system and metabolism regulation. Under malnutrition state, leptin acts as an immunosuppressive factor [42].

Advertisement

6. Leptin as a pro-angiogenic factor

In 1998, Sierra-Hongmans reported that vascular endothelial cells express leptin receptors, especially the long-form. This discovery leads to an insight into the role of leptin in angiogenesis [57]. The angiogenic impact of leptin was conformed used on in vitro and in vivo models analysis [3, 58]. Jin et al. proved that leptin could induce angiogenesis in the cornea of the Zucker obese rat model through the activation of the Ob-R gene [59]. Leptin exerts a paracrine mode of action in tissues and activates various signaling during the promotion of angiogenesis. This endocrine hormone activates Akt signaling pathway and mediates NO-induced vasodilation [60]. In endothelial cell migration, leptin signals through the ERK pathway and activates the PI3k, Akt, and eNOS molecules. By stimulating the local neovascularization in adipose tissue, leptin promotes its release into the vascular system. This process enhances fatty acid oxidation and supports maintaining a proper balance between adipose tissue’s fat deposits and blood supply [61]. Even though the vascular fenestration capacity of leptin is poorly understood, the effect is found similar to VEGF [62]. Leptin plays a crucial role in exchanging nutrients between the fetus and maternal circulation in the placenta via enhancing vascular permeability and could induce angiogenesis in the placenta [61].

Advertisement

7. Leptin and pathogenesis

Leptin does not only imply energy homeostasis but also extends its regulatory function at infectious conditions. But the contagious status regulation mode of leptin is the least explored signaling mechanism. Latest research support that leptin could activate phagocytosis of macrophages and could secure the immune cells from pathogenic infections [63]. In Klebsiella pneumonia infection, exogenous administration of leptin shows CD11b dependent phagocytosis [64]. It protects lymphocyte deficient mice from various conditions [65]. Several studies have strongly highlighted the therapeutic application of the molecule to innervate infectious diseases, including AIDS [66].

Advertisement

8. Leptin resistance with disease

However, under certain conditions, like obesity, leptin levels decrease in association with leptin resistance. But it is still unclear how the leptin resistance mechanism is exerted throughout the tissue. So far, studies have suggested leptin resistance with metabolic process and revealed a defect in the Ob-R leptin receptor gene [67]. Up to date, the leptin resistance mechanism has been categorized as follows: gene mutation specific to the leptin structure, defect in the transport of leptin through the blood–brain barrier, and malfunctions of leptin receptors. Among these, mutations are rare in humans, occurring in substitution of guanine by adenine at the donor splice site of exon 16 of the leptin gene [68]. Second, the brain’s blood vessels usually express leptin receptors and transport leptin into the cerebrospinal fluid. But excessive levels of leptin in the bloodstream decrease the permeability of BBB, thus develops leptin resistance [69]. Finally, the serum level of leptin significantly affects the transcriptional level of the OB (ob) gene and the equilibrium of leptin secretion in adipose tissue. In such cases, these dramatic changes promote leptin resistance until leptin level remains standard in the bloodstream. These changes have been widely observed in obesity [70].

Furthermore, several stimuli affect leptin resistance, including the circadian cycle. Interestingly, leptin also develops its leptin resistance, observed in diet-induced obesity [71]. This leptin resistance also provides an environment for the accumulation of immune response against pathogens, particularly high-fat diet-induced inflammation, which activates inflammatory cytokines [72]. But, in-depth leptin resistance mechanisms need much more attention.

Advertisement

9. Leptin role in disease conditions

9.1 Metabolic syndrome and obesity

Fat tissue is an energy storage tissue that functions as a negative feedback loop in energy homeostasis [73]. Homozygous mutation of leptin causes extreme obesity, diabetes and suppresses glucose metabolism in insulin-deficient diabetes [8]. The ob/ob mice model has relatively higher food intake and observed a larger volume of lipid accumulation in the liver than the control group [74]. It has been assumed that nearly 95% of individuals have resistance against leptin [75]. The type 2 diabetes condition is noted with an increased level of leptin and suggested using leptin as a biomarker to study the effect of obesity in diabetes-related morbidities [76]. Some studies also reported that higher leptin levels are associated with the risk of heart-related problems in obese individuals [76, 77]. In younger adults, elevated leptin levels are positively correlated with HOMA-IR and BMI index [78].

Development of severe early-onset obesity and hyperphagia are common in people with homozygous LEP mutation [79]. Replacement of leptin from a therapeutic viewpoint has improved insulin sensitivity and thus proved the role of leptin in metabolic disorders, including T2DM. In humans, serum leptin level is positively correlated with the percentage of body fat, fat mass, size of adipocytes, and BMI [80]. Obesity connected with the enlargement of adipose cells enhances the serum leptin level, which further results in the progression of chronic hyperinsulinemia. The majority of obese patients are hyper leptinaemic which supports the development of hypertension, metabolic syndrome, and cardiovascular diseases [81]. Mutation in the leptin receptor located at the hypothalamus alters the transport of leptin across the blood–brain barrier. This incidence increases the level of serum leptin and hence diet-induced obesity. Obesity connected with the leptin receptor mutation is linked with insulin resistance and in the development of T2DM [82].

9.2 Cardiovascular diseases

The level of leptin could influence the function of the heart. It could lead to the progression of many heart-related problems such as coronary artery disease, stroke, chronic kidney disease (CKD), peripheral artery disease (DAP), carotid plaque instability [83]. It was observed that elevated level of serum leptin in obese patients contributes to the low-grade systemic inflammation in favor to develop cardiovascular disease. Moreover, a high level of leptin is used as a biomarker to measure the progression of heart failure in patients with dilated cardiomyopathy [84]. On the other hand, many studies using rodent, obese and diabetic models highlighted the beneficial impact of leptin on cardiac metabolism through glucose metabolism and fatty acid oxidation. This evidence suggested that leptin compensates for cardiac insults due to ischemia and heart failure [85]. Leptin signaling in the modulation of heart function is studied extensively using animal models. These studies demonstrated that impaired cardiac leptin signaling majorly reflects in metabolic inflexibility for glucose utilization, defects in cardiac contractibility, impaired recovery of cardiac function due to coronary artery ligation [86, 87]. Clinical data cemented that plasma leptin levels are associated with LV hypertrophy and increased myocardial wall thickening [88]. Leptin also increased the blood pressure level in obese individuals with a loss-of-function mutation in leptin or leptin receptor [89]. Thus, a leptin-mediated increase in blood pressure directly increases the heartbeat rate, developing into cardiac hypertrophy through the sympathetic nervous mechanism [90].

Leptin-mediated aldosterone synthesis impairs myocardial relaxation and contributes to cardiovascular diseases through a novel mechanism associated with endothelial dysfunctions [91]. Increased plasma leptin levels positively correlate with the number of stenotic coronary arteries in patients with coronary artery disease [92]. In vitro analysis using HUVEC cells demonstrated that leptin induces chronic oxidative stress in ECs and contributes to vascular pathology development [93]. Also, the cytokine hormone leptin could stimulate vascular smooth muscle cells proliferation and migration, thereby increasing calcification and vascular lesions [94]. Altogether, it was suggested that hypertension, obesity, and endothelial dysfunctions are more frequent in T2DM patients with elevated leptin levels [95].

9.3 Tumor progression

Cancer progression is a complex process that includes the interaction between ECs, fibroblast, insusceptible cells, and adipocytes [96]. Normal epithelial cells do not express leptin and leptin receptors but are overexpressed in a cancerous environment. Leptin enhances the survival rate of cancer cells through the activation of a downstream signaling molecule known as sirtuin-dependent NAD-dependent deacetylase 1 (SIRT 1) [97]. Leptin can activate many signaling pathways in cancer directly by activating TNF alpha, IL-6, ROS, VEGF, MMP2, and MMP9. It can also support tumor growth by activating JAK/STAT, Akt, FGF2, and NO molecules through receptor (Ob-R) binding mechanisms in ECs [98, 99]. The appetite hormone can potentially interact with pre-neoplastic or cancerous breast epithelium in a breast cancer environment. Leptin secreted by the breast cancer surroundings inhibits inflammatory cytokines and thus blocks macrophages’ production [100, 101]. The cytokine enhances neovascularization through VEGF in many cancerous conditions [102, 103].

Increased levels of serum leptin and insulin under obese conditions cause colorectal cancer [104]. Leptin supports the proliferation and invasiveness of colonic cells. Leptin receptors are found to express in human colon cell lines and are believed to initiate cancer angiogenesis. Hyperlipidemia and insulin resistance can cause low-grade systemic inflammation that promotes proliferation and angiogenesis and inhibits apoptotic rate in colon cancer [105]. Leptin and its receptors express in papillary thyroid tumors and enhance the pathogenicity through PI3K/Akt pathway [106].

Obesity enhances the concentration of leptin around the pancreatic carcinogenic environment. The enhanced concentration of leptin favors vascularization, migration, and invasiveness of pancreatic tumor cells [105]. Leptin has a crucial role in developing the non-alcoholic fatty liver disease (NAFLD) via insulin resistance. This imbalance ultimately worsens hepatic inflammation and results in the development of liver fibrosis [107]. The receptor Ob-R identified in Kupffer cells (KC), and binding of leptin with receptor enhances the expression of TGF beta, TIMP1 in liver fibrosis scenario [108]. However, the direct role of leptin in liver cancer is controversial, with some reports suggesting its role in liver cancer. In contrast, others offer its inhibitory potential on tumor size in hepatic cancer [109, 110]. The level of leptin was found to decrease in patients with cancer cachexia compared to non-cancer cachexia [111].

Advertisement

10. Leptin therapeutics past and future

The significant need for clinical implications of leptin is to regulate the regular physiological role of leptin in pathological conditions. There is a correlation between body weight loss and serum levels of leptin. As a result, several therapeutic approaches have been implemented for the use of leptin in obesity control. However, increased resistance to leptin is also a significant issue in the treatment of obesity. But, a combination of therapeutic approaches may be helpful to these problems [112, 113].

Among the adipocyte secreted hormones, leptin is the front that has been used for the treatment of hypoleptinemia status clinically. The most important therapeutic benefits of leptin are rely on providing a novel method for treating the conditions connected with mutation of leptin gene and lipodystrophy in humans [114]. Treatment with exogenous leptin in obese patients concluded that leptin can decrease the body weight and fat tissue of the subjects [115]. It was also noted that leptin excerpts a dose-dependent regulating potential as individuals energy intake and appetite [116, 117]. Development of leptin analogous with full biological effect, especially with the potential to cross the blood–brain barrier, can improve the results obtained from leptin therapy focusing on obesity management. The administration of leptin can accelerate wound healing in diabetic ob/ob and wild-type mice in a dose-dependent manner through leptin receptor mediation [118].

Exogenous administration of leptin can regulate fatty acid oxidation in muscles and control triglyceride synthesis in the liver [119, 120]. Even though the mechanisms in humans are not clear, administration of leptin and adiponectin was found to improve insulin resistance in type 2 diabetic conditions [121, 122]. The immune-modulatory impact of exogenous leptin administration in rodents highlighted that the cytokine could activate encephalomyelitis [123]. Various in vitro assays also supported the immune stimulator action of leptin [124, 125]. Identifying high-affinity-binding molecules to control the level of circulating leptin is suggested as an advanced therapy for treating arthritis and inflammatory bowel disease. In addition, replacement with recombinant methionyl human leptin is a brilliant choice for treating pathological conditions associated with relative or absolute leptin deficiency and restoring immune functions [126]. One of the future therapeutic approaches of leptin relies on its use as a natural adjuvant in vaccinations since it can stimulate T helper I responses while down-modulating regulatory T cells [127].

Considering the cancerous conditions, ATLO-ACA, an Ob-R antagonist peptide, finds effective for treating triple-negative breast cancers in experimental models [128]. Also, therapy based on leptin/Ob R axis function inhibition was identified as adjunctive therapy for newly diagnosed and recurrent glioblastoma [129]. The modern therapeutic approaches of leptin are connected with molecular approaches at gene levels are 1) CRISPR-Cas 9 connected with floxed leptin-locus based approaches - to lower the leptin levels, 2) Cre-lox P- generation of one copy of Lep eliminates – to lower the leptin levels, glucose and insulin tolerance, c) administration of neutralizing leptin-specific antibodies –to reduce the circulating levels of leptin to reduce food intake and hepatic stenosis [130]. Administration of human recombinant leptin accelareated dose-dependent sprouting angiogenesis and hypothesized that the application of human recombinant leptin could improve the wound healing process through neovascularization [3]. So far, evidence gathered from previous studies highlights the role of leptin in therapeutic applications. But overcoming leptin resistance is a significant challenge in leptin-based therapy.

11. Conclusion

In conclusion, leptin is considered an essential pleiotropic adipokine with various effects on biological systems. However, leptin’s structural and functional characteristics and its receptors are characterized by a unique signaling mechanism. Focusing on leptin could be a therapeutic approach to manage autoimmune inflammation associated with obesity, cancer, and metabolic diseases. But further research is needed to understand the relationship between leptin on biological systems, as it has complex signaling mechanisms.

Acknowledgments

Financial support from the Science and Engineering Research Board (DST-SERB), Government of India (Project file no: EEQ/2019/000568) is greatly acknowledged.

Conflicts of interest

The authors declare no conflict of interest.

Authors Contribution

DK collected information and prepared the first draft, RM and JS critically evaluated and coordinated with final draft preparation.

References

  1. 1. Ramos-Lobo AM, Donato J Jr. The role of leptin in health and disease. Temperature (Austin). 2017 May 26;4(3):258-291. DOI: 10.1080/23328940.2017.1327003. PMID: 28944270; PMCID: PMC5605162
  2. 2. Pralong FP, Gaillard RC. Neuroendocrine effects of leptin. Pituitary. 2001 Jan-Apr;4(1-2):25-32. DOI: 10.1023/a:1012930609563. PMID: 11824505
  3. 3. Manjunathan R, Ragunathan M. In ovo administration of human recombinant leptin shows dose-dependent angiogenic effect on chicken chorioallantoic membrane. Biol Res. 2015 Jun 10;48(1):29. DOI: 10.1186/s40659-015-0021-z. PMID: 26060038; PMCID: PMC4470073
  4. 4. Manjunathan R, Devarajan N, Ragunathan M. Possible Mechanism of Human Recombinant Leptin-Induced VEGF A Synthesis via PI3K/Akt/mTOR/S6 Kinase Signaling Pathway while Inducing Angiogenesis: An Analysis Using Chicken Chorioallantoic Membrane Model. J Vasc Res. 2021 Jun 24:1-18. DOI: 10.1159/000516498. Epub ahead of print. PMID: 34167108
  5. 5. Lee GH, Proenca R, Montez JM, Carroll KM, Darvishzadeh JG, Lee JI, Friedman JM. Abnormal splicing of the leptin receptor in diabetic mice. Nature. 1996 Feb 15;379(6566):632-635. DOI: 10.1038/379632a0. PMID: 8628397
  6. 6. Vázquez-Del Mercado M, Martínez-García EA. Leptin as an open secret in the physiopathology of rheumatic diseases. Clin Rheumatol. 2020 Feb;39(2):301-303. DOI: 10.1007/s10067-019-04908-4. Epub 2020 Jan 11. PMID: 31927713
  7. 7. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest. 2003 May;111(9):1409-21. DOI: 10.1172/JCI17490. PMID: 12727933; PMCID: PMC154448
  8. 8. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994 Dec 1;372(6505):425-32. DOI: 10.1038/372425a0. Erratum in: Nature 1995 Mar 30;374(6521):479. PMID: 7984236
  9. 9. Chung WK, Power-Kehoe L, Chua M, Leibel RL. Mapping of the OB receptor to 1p in a region of nonconserved gene order from mouse and rat to human. Genome Res. 1996 May;6(5):431-438. DOI: 10.1101/gr.6.5.431. PMID: 8743992
  10. 10. Campfield LA, Smith FJ, Burn P. The OB protein (leptin) pathway--a link between adipose tissue mass and central neural networks. Horm Metab Res. 1996 Dec;28(12):619-632. DOI: 10.1055/s-2007-979867. PMID: 9013731
  11. 11. Bjørbaek C, Uotani S, da Silva B, Flier JS. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J Biol Chem. 1997 Dec 19;272(51):32686-32695. DOI: 10.1074/jbc.272.51.32686. PMID: 9405487
  12. 12. Levine JA, Eberhardt NL, Jensen MD. Leptin responses to overfeeding: relationship with body fat and nonexercise activity thermogenesis. J Clin Endocrinol Metab. 1999 Aug;84(8):2751-2754. DOI: 10.1210/jcem.84.8.5910. PMID: 10443673
  13. 13. stlund Jr, R., Yang, J. W., Klein, S. & Gingerich, R. Relation between plasma leptin concentration and body fat, gender, diet, age, and metabolic covariates. The journal of clinical endocrinology & metabolism81, 3909-3913 (1996)
  14. 14. Licinio J, Mantzoros C, Negrão AB, Cizza G, Wong ML, Bongiorno PB, Chrousos GP, Karp B, Allen C, Flier JS, Gold PW. Human leptin levels are pulsatile and inversely related to pituitary-adrenal function. Nat Med. 1997 May;3(5):575-579. DOI: 10.1038/nm0597-575. PMID: 9142131
  15. 15. Jiang L, You J, Yu X, Gonzalez L, Yu Y, Wang Q, Yang G, Li W, Li C, Liu Y. Tyrosine-dependent and -independent actions of leptin receptor in control of energy balance and glucose homeostasis. Proc Natl Acad Sci U S A. 2008 Nov 25;105(47):18619-24. DOI: 10.1073/pnas.0804589105. Epub 2008 Nov 17. PMID: 19015522; PMCID: PMC2587560
  16. 16. Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, Neel BG, Schwartz MW, Myers MG Jr. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature. 2003 Feb 20;421(6925):856-859. DOI: 10.1038/nature01388. PMID: 12594516
  17. 17. Varela L, Horvath TL. Leptin and insulin pathways in POMC and AgRP neurons that modulate energy balance and glucose homeostasis. EMBO Rep. 2012 Dec;13(12):1079-86. DOI: 10.1038/embor.2012.174. Epub 2012 Nov 13. PMID: 23146889; PMCID: PMC3512417
  18. 18. Rahmouni K, Sigmund CD, Haynes WG, Mark AL. Hypothalamic ERK mediates the anorectic and thermogenic sympathetic effects of leptin. Diabetes. 2009 Mar;58(3):536-42. DOI: 10.2337/db08-0822. Epub 2008 Dec 9. PMID: 19066310; PMCID: PMC2646051
  19. 19. Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, Mu J, Foufelle F, Ferré P, Birnbaum MJ, Stuck BJ, Kahn BB. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004 Apr 1;428(6982):569-574. DOI: 10.1038/nature02440. PMID: 15058305
  20. 20. Minokoshi Y, Kim YB, Peroni OD, Fryer LG, Müller C, Carling D, Kahn BB. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature. 2002 Jan 17;415(6869):339-343. DOI: 10.1038/415339a. PMID: 11797013
  21. 21. Paz-Filho G, Mastronardi C, Franco CB, Wang KB, Wong ML, Licinio J. Leptin: molecular mechanisms, systemic pro-inflammatory effects, and clinical implications. Arq Bras Endocrinol Metabol. 2012 Dec;56(9):597-607. DOI: 10.1590/s0004-27302012000900001. PMID: 23329181
  22. 22. Iikuni N, Lam QL, Lu L, Matarese G, La Cava A. Leptin and Inflammation. Curr Immunol Rev. 2008 May 1;4(2):70-79. DOI: 10.2174/157339508784325046. PMID: 20198122; PMCID: PMC2829991
  23. 23. Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med. 2012 Feb;33(1):35-45. DOI: 10.1016/j.mam.2011.10.012. Epub 2011 Oct 21. PMID: 22040697
  24. 24. Frühbeck G, Gómez-Ambrosi J. Modulation of the leptin-induced white adipose tissue lipolysis by nitric oxide. Cell Signal. 2001 Nov;13(11):827-833. DOI: 10.1016/s0898-6568(01)00211-x. PMID: 11583918
  25. 25. Frühbeck G. A heliocentric view of leptin. Proc Nutr Soc. 2001 Aug;60(3):301-318. DOI: 10.1079/pns200196. PMID: 11681805
  26. 26. Hardwick JC, Van Den Brink GR, Offerhaus GJ, Van Deventer SJ, Peppelenbosch MP. Leptin is a growth factor for colonic epithelial cells. Gastroenterology. 2001 Jul;121(1):79-90. DOI: 10.1053/gast.2001.25490. PMID: 11438496
  27. 27. Islam MS, Morton NM, Hansson A, Emilsson V. Rat insulinoma-derived pancreatic beta-cells express a functional leptin receptor that mediates a proliferative response. Biochem Biophys Res Commun. 1997 Sep 29;238(3):851-855. DOI: 10.1006/bbrc.1997.7399. PMID: 9325180
  28. 28. Sweeney G. Leptin signalling. Cell Signal. 2002 Aug;14(8):655-663. DOI: 10.1016/s0898-6568(02)00006-2. PMID: 12020765
  29. 29. Guyenet SJ, Schwartz MW. Clinical review: Regulation of food intake, energy balance, and body fat mass: implications for the pathogenesis and treatment of obesity. J Clin Endocrinol Metab. 2012 Mar;97(3):745-55. DOI: 10.1210/jc.2011-2525. Epub 2012 Jan 11. PMID: 22238401; PMCID: PMC3319208
  30. 30. Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006 Sep 21;443(7109):289-295. DOI: 10.1038/nature05026. PMID: 16988703
  31. 31. Picó C, Oliver P, Sánchez J, Palou A. Gastric leptin: a putative role in the short-term regulation of food intake. Br J Nutr. 2003 Oct;90(4):735-741. DOI: 10.1079/bjn2003945. PMID: 13129441
  32. 32. Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest. 2003 May;111(9):1409-21. DOI: 10.1172/JCI17490. PMID: 12727933; PMCID: PMC154448
  33. 33. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, Kim S, Lallone R, Ranganathan S, et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995 Nov;1(11):1155-1161. DOI: 10.1038/nm1195-1155. PMID: 7584987
  34. 34. Münzberg H, Flier JS, Bjørbaek C. Region-specific leptin resistance within the hypothalamus of diet-induced obese mice. Endocrinology. 2004 Nov;145(11):4880-4889. DOI: 10.1210/en.2004-0726. Epub 2004 Jul 22. PMID: 15271881
  35. 35. Igel M, Becker W, Herberg L, Joost HG. Hyperleptinemia, leptin resistance, and polymorphic leptin receptor in the New Zealand obese mouse. Endocrinology. 1997 Oct;138(10):4234-4239. DOI: 10.1210/endo.138.10.5428. PMID: 9322935
  36. 36. Zhang Y, Dallner OS, Nakadai T, Fayzikhodjaeva G, Lu YH, Lazar MA, Roeder RG, Friedman JM. A noncanonical PPARγ/RXRα-binding sequence regulates leptin expression in response to changes in adipose tissue mass. Proc Natl Acad Sci U S A. 2018 Jun 26;115(26):E6039-E6047. DOI: 10.1073/pnas.1806366115. Epub 2018 Jun 11. PMID: 29891714; PMCID: PMC6042069
  37. 37. Dallner OS, Marinis JM, Lu YH, Birsoy K, Werner E, Fayzikhodjaeva G, Dill BD, Molina H, Moscati A, Kutalik Z, Marques-Vidal P, Kilpeläinen TO, Grarup N, Linneberg A, Zhang Y, Vaughan R, Loos RJF, Lazar MA, Friedman JM. Dysregulation of a long noncoding RNA reduces leptin leading to a leptin-responsive form of obesity. Nat Med. 2019 Mar;25(3):507-516. DOI: 10.1038/s41591-019-0370-1. Epub 2019 Mar 6. PMID: 30842678
  38. 38. Derghal A, Djelloul M, Azzarelli M, Degonon S, Tourniaire F, Landrier JF, Mounien L. MicroRNAs are involved in the hypothalamic leptin sensitivity. Epigenetics. 2018;13(10-11):1127-1140. DOI: 10.1080/15592294.2018.1543507. Epub 2018 Nov 11. PMID: 30395773; PMCID: PMC6342074
  39. 39. Kiernan K, MacIver NJ. The Role of the Adipokine Leptin in Immune Cell Function in Health and Disease. Front Immunol. 2021 Jan 29;11:622468. DOI: 10.3389/fimmu.2020.622468. PMID: 33584724; PMCID: PMC7878386
  40. 40. Gerriets VA, Danzaki K, Kishton RJ, Eisner W, Nichols AG, Saucillo DC, Shinohara ML, MacIver NJ. Leptin directly promotes T-cell glycolytic metabolism to drive effector T-cell differentiation in a mouse model of autoimmunity. Eur J Immunol. 2016 Aug;46(8):1970-83. DOI: 10.1002/eji.201545861. Epub 2016 Jun 13. PMID: 27222115; PMCID: PMC5154618
  41. 41. Busso N, So A, Chobaz-Péclat V, Morard C, Martinez-Soria E, Talabot-Ayer D, Gabay C. Leptin signaling deficiency impairs humoral and cellular immune responses and attenuates experimental arthritis. J Immunol. 2002 Jan 15;168(2):875-882. DOI: 10.4049/jimmunol.168.2.875. PMID: 11777985
  42. 42. Pérez-Pérez A, Vilariño-García T, Fernández-Riejos P, Martín-González J, Segura-Egea JJ, Sánchez-Margalet V. Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev. 2017 Jun;35:71-84. DOI: 10.1016/j.cytogfr.2017.03.001. Epub 2017 Mar 4. PMID: 28285098
  43. 43. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI. Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression. Nature. 1998 Aug 27;394(6696):897-901. DOI: 10.1038/29795. PMID: 9732873
  44. 44. Saucillo DC, Gerriets VA, Sheng J, Rathmell JC, Maciver NJ. Leptin metabolically licenses T cells for activation to link nutrition and immunity. J Immunol. 2014 Jan 1;192(1):136-44. DOI: 10.4049/jimmunol.1301158. Epub 2013 Nov 22. PMID: 24273001; PMCID: PMC3872216
  45. 45. Su X, Cheng Y, Chang D. The Important Role of Leptin in Modulating the Risk of Dermatological Diseases. Front Immunol. 2021 Feb 1;11:593564. DOI: 10.3389/fimmu.2020.593564. PMID: 33597945; PMCID: PMC7882601
  46. 46. Grosfeld A, Turban S, André J, Cauzac M, Challier JC, Hauguel-de Mouzon S, Guerre-Millo M. Transcriptional effect of hypoxia on placental leptin. FEBS Lett. 2001 Aug 3;502(3):122-126. DOI: 10.1016/s0014-5793(01)02673-4. PMID: 11583112
  47. 47. Bouloumie A, Marumo T, Lafontan M, Busse R. Leptin induces oxidative stress in human endothelial cells. FASEB J. 1999 Jul;13(10):1231-1238. PMID: 10385613
  48. 48. Gainsford T, Willson TA, Metcalf D, Handman E, McFarlane C, Ng A, Nicola NA, Alexander WS, Hilton DJ. Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells. Proc Natl Acad Sci U S A. 1996 Dec 10;93(25):14564-8. DOI: 10.1073/pnas.93.25.14564. PMID: 8962092; PMCID: PMC26173
  49. 49. Janik JE, Curti BD, Considine RV, Rager HC, Powers GC, Alvord WG, Smith JW 2nd, Gause BL, Kopp WC. Interleukin 1 alpha increases serum leptin concentrations in humans. J Clin Endocrinol Metab. 1997 Sep;82(9):3084-3086. DOI: 10.1210/jcem.82.9.4214. PMID: 9284748
  50. 50. Conus S, Bruno A, Simon HU. Leptin is an eosinophil survival factor. J Allergy Clin Immunol. 2005 Dec;116(6):1228-1234. DOI: 10.1016/j.jaci.2005.09.003. Epub 2005 Oct 24. PMID: 16337450
  51. 51. Ünlü B, Türsen Ü. Autoimmune skin diseases and the metabolic syndrome. Clin Dermatol. 2018 Jan-Feb;36(1):67-71. DOI: 10.1016/j.clindermatol.2017.09.012. Epub 2017 Sep 8. PMID: 29241755
  52. 52. Deng T, Lyon CJ, Bergin S, Caligiuri MA, Hsueh WA. Obesity, Inflammation, and Cancer. Annu Rev Pathol. 2016 May 23;11:421-449. DOI: 10.1146/annurev-pathol-012615-044359. PMID: 27193454
  53. 53. Hirt PA, Castillo DE, Yosipovitch G, Keri JE. Skin changes in the obese patient. J Am Acad Dermatol. 2019 Nov;81(5):1037-1057. DOI: 10.1016/j.jaad.2018.12.070. PMID: 31610857
  54. 54. Buttgereit F, Burmester GR, Brand MD. Bioenergetics of immune functions: fundamental and therapeutic aspects. Immunol Today. 2000 Apr;21(4):192-199. DOI: 10.1016/s0167-5699(00)01593-0. PMID: 10740243
  55. 55. Lee FY, Li Y, Yang EK, Yang SQ, Lin HZ, Trush MA, Dannenberg AJ, Diehl AM. Phenotypic abnormalities in macrophages from leptin-deficient, obese mice. Am J Physiol. 1999 Feb;276(2):C386-C394. DOI: 10.1152/ajpcell.1999.276.2.C386. PMID: 9950766
  56. 56. Zarkesh-Esfahani H, Pockley G, Metcalfe RA, Bidlingmaier M, Wu Z, Ajami A, Weetman AP, Strasburger CJ, Ross RJ. High-dose leptin activates human leukocytes via receptor expression on monocytes. J Immunol. 2001 Oct 15;167(8):4593-4599. DOI: 10.4049/jimmunol.167.8.4593. PMID: 11591788
  57. 57. Sierra-Honigmann MR, Nath AK, Murakami C, García-Cardeña G, Papapetropoulos A, Sessa WC, Madge LA, Schechner JS, Schwabb MB, Polverini PJ, Flores-Riveros JR. Biological action of leptin as an angiogenic factor. Science. 1998 Sep 11;281(5383):1683-1686. DOI: 10.1126/science.281.5383.1683. PMID: 9733517
  58. 58. Bouloumié A, Drexler HC, Lafontan M, Busse R. Leptin, the product of Ob gene, promotes angiogenesis. Circ Res. 1998 Nov 16;83(10):1059-1066. DOI: 10.1161/01.res.83.10.1059. PMID: 9815153
  59. 59. Jin X, Fukuda N, Su J, Takagi H, Lai Y, Lin Z, Kanmatsuse K, Wang ZW, Unger RH. Effects of leptin on endothelial function with OB-Rb gene transfer in Zucker fatty rats. Atherosclerosis. 2003 Aug;169(2):225-233. DOI: 10.1016/s0021-9150(03)00159-x. PMID: 12921973
  60. 60. Vecchione C, Maffei A, Colella S, Aretini A, Poulet R, Frati G, Gentile MT, Fratta L, Trimarco V, Trimarco B, Lembo G. Leptin effect on endothelial nitric oxide is mediated through Akt-endothelial nitric oxide synthase phosphorylation pathway. Diabetes. 2002 Jan;51(1):168-173. DOI: 10.2337/diabetes.51.1.168. PMID: 11756337
  61. 61. Cao R, Brakenhielm E, Wahlestedt C, Thyberg J, Cao Y. Leptin induces vascular permeability and synergistically stimulates angiogenesis with FGF-2 and VEGF. Proc Natl Acad Sci U S A. 2001 May 22;98(11):6390-5. DOI: 10.1073/pnas.101564798. Epub 2001 May 8. PMID: 11344271; PMCID: PMC33478
  62. 62. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 1983 Feb 25;219(4587):983-985. DOI: 10.1126/science.6823562. PMID: 6823562
  63. 63. Bruno A, Conus S, Schmid I, Simon HU. Apoptotic pathways are inhibited by leptin receptor activation in neutrophils. J Immunol. 2005 Jun 15;174(12):8090-8096. DOI: 10.4049/jimmunol.174.12.8090. PMID: 15944317
  64. 64. Moore SI, Huffnagle GB, Chen GH, White ES, Mancuso P. Leptin modulates neutrophil phagocytosis of Klebsiella pneumoniae. Infect Immun. 2003 Jul;71(7):4182-5. DOI: 10.1128/IAI.71.7.4182-4185.2003. PMID: 12819114; PMCID: PMC161963
  65. 65. Merrick JC, Edelson BT, Bhardwaj V, Swanson PE, Unanue ER. Lymphocyte apoptosis during early phase of Listeria infection in mice. Am J Pathol. 1997 Sep;151(3):785-92. PMID: 9284827; PMCID: PMC1857845
  66. 66. Coffey MJ, Phare SM, Kazanjian PH, Peters-Golden M. 5-Lipoxygenase metabolism in alveolar macrophages from subjects infected with the human immunodeficiency virus. J Immunol. 1996 Jul 1;157(1):393-399. PMID: 8683143
  67. 67. Malli F, Papaioannou AI, Gourgoulianis KI, Daniil Z. The role of leptin in the respiratory system: an overview. Respir Res. 2010 Oct 31;11(1):152. DOI: 10.1186/1465-9921-11-152. PMID: 21040518; PMCID: PMC2988727
  68. 68. Wabitsch M, Funcke JB, Lennerz B, Kuhnle-Krahl U, Lahr G, Debatin KM, Vatter P, Gierschik P, Moepps B, Fischer-Posovszky P. Biologically inactive leptin and early-onset extreme obesity. N Engl J Med. 2015 Jan 1;372(1):48-54. DOI: 10.1056/NEJMoa1406653. PMID: 25551525
  69. 69. Mantzoros CS. The role of leptin in human obesity and disease: a review of current evidence. Ann Intern Med. 1999 Apr 20;130(8):671-680. DOI: 10.7326/0003-4819-130-8-199904200-00014. PMID: 10215564.
  70. 70. Mainardi M, Pizzorusso T, Maffei M. Environment, leptin sensitivity, and hypothalamic plasticity. Neural Plast. 2013;2013:438072. DOI: 10.1155/2013/438072. Epub 2013 Jul 18. PMID: 23970977; PMCID: PMC3732608
  71. 71. Gonzalez-Carter D, Goode AE, Fiammengo R, Dunlop IE, Dexter DT, Porter AE. Inhibition of Leptin-ObR Interaction Does not Prevent Leptin Translocation Across a Human Blood-Brain Barrier Model. J Neuroendocrinol. 2016 Jun;28(6). DOI: 10.1111/jne.12392. PMID: 27037668
  72. 72. Mattace Raso G, Simeoli R, Russo R, Iacono A, Santoro A, Paciello O, Ferrante MC, Canani RB, Calignano A, Meli R. Effects of sodium butyrate and its synthetic amide derivative on liver inflammation and glucose tolerance in an animal model of steatosis induced by high fat diet. PLoS One. 2013 Jul 5;8(7):e68626. DOI: 10.1371/journal.pone.0068626. PMID: 23861927; PMCID: PMC3702592
  73. 73. Kelesidis T, Kelesidis I, Chou S, Mantzoros CS. Narrative review: the role of leptin in human physiology: emerging clinical applications. Ann Intern Med. 2010 Jan 19;152(2):93-100. DOI: 10.7326/0003-4819-152-2-201001190-00008. PMID: 20083828; PMCID: PMC2829242
  74. 74. Minokoshi Y, Toda C, Okamoto S. Regulatory role of leptin in glucose and lipid metabolism in skeletal muscle. Indian J Endocrinol Metab. 2012 Dec;16(Suppl 3):S562-8. DOI: 10.4103/2230-8210.105573. PMID: 23565491; PMCID: PMC3602985
  75. 75. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, Kim S, Lallone R, Ranganathan S, et al. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nat Med. 1995 Nov;1(11):1155-1161. DOI: 10.1038/nm1195-1155. PMID: 7584987
  76. 76. Ahima RS. Central actions of adipocyte hormones. Trends Endocrinol Metab. 2005 Sep;16(7):307-313. DOI: 10.1016/j.tem.2005.07.010. PMID: 16098759
  77. 77. igneshwaran B, Wahal A, Aggarwal S, Priyadarshini P, Bhattacharjee H, Khadgawat R, Yadav R. Impact of Sleeve Gastrectomy on Type 2 Diabetes Mellitus, Gastric Emptying Time, Glucagon-Like Peptide 1 (GLP-1), Ghrelin and Leptin in Non-morbidly Obese Subjects with BMI 30-35.0 kg/m2: a Prospective Study. Obes Surg. 2016 Dec;26(12):2817-2823. DOI: 10.1007/s11695-016-2226-9. PMID: 27185177
  78. 78. Belhayara MI, Mellouk Z, Hamdaoui MS, Bachaoui M, Kheroua O, Malaisse WJ. Relationship between the insulin resistance and circulating predictive biochemical markers in metabolic syndrome among young adults in western Algeria. Diabetes Metab Syndr. 2019 Jan-Feb;13(1):504-509. DOI: 10.1016/j.dsx.2018.11.019. Epub 2018 Nov 3. PMID: 30641755
  79. 79. Dodd GT, Worth AA, Nunn N, Korpal AK, Bechtold DA, Allison MB, Myers MG Jr, Statnick MA, Luckman SM. The thermogenic effect of leptin is dependent on a distinct population of prolactin-releasing peptide neurons in the dorsomedial hypothalamus. Cell Metab. 2014 Oct 7;20(4):639-49. DOI: 10.1016/j.cmet.2014.07.022. Epub 2014 Aug 28. PMID: 25176149; PMCID: PMC4192552
  80. 80. Elmquist JK, Ahima RS, Maratos-Flier E, Flier JS, Saper CB. Leptin activates neurons in ventrobasal hypothalamus and brainstem. Endocrinology. 1997 Feb;138(2):839-842. DOI: 10.1210/endo.138.2.5033. PMID: 9003024
  81. 81. Fischer AW, Hoefig CS, Abreu-Vieira G, de Jong JMA, Petrovic N, Mittag J, Cannon B, Nedergaard J. Leptin Raises Defended Body Temperature without Activating Thermogenesis. Cell Rep. 2016 Feb 23;14(7):1621-1631. DOI: 10.1016/j.celrep.2016.01.041. Epub 2016 Feb 11. PMID: 26876182
  82. 82. Garfield AS, Li C, Madara JC, Shah BP, Webber E, Steger JS, Campbell JN, Gavrilova O, Lee CE, Olson DP, Elmquist JK, Tannous BA, Krashes MJ, Lowell BB. A neural basis for melanocortin-4 receptor-regulated appetite. Nat Neurosci. 2015 Jun;18(6):863-71. DOI: 10.1038/nn.4011. Epub 2015 Apr 27. PMID: 25915476; PMCID: PMC4446192
  83. 83. Athyros VG, Katsiki N, Karagiannis A, Mikhailidis DP. Editorial: should chronic kidney disease be considered as a coronary heart disease equivalent? Curr Vasc Pharmacol. 2012 May;10(3):374-377. DOI: 10.2174/157016112799959422. PMID: 22272896
  84. 84. Levine JA, Eberhardt NL, Jensen MD. Role of nonexercise activity thermogenesis in resistance to fat gain in humans. Science. 1999 Jan 8;283(5399):212-214. DOI: 10.1126/science.283.5399.212. PMID: 9880251
  85. 85. Lute B, Jou W, Lateef DM, Goldgof M, Xiao C, Piñol RA, Kravitz AV, Miller NR, Huang YG, Girardet C, Butler AA, Gavrilova O, Reitman ML. Biphasic effect of melanocortin agonists on metabolic rate and body temperature. Cell Metab. 2014 Aug 5;20(2):333-45. DOI: 10.1016/j.cmet.2014.05.021. Epub 2014 Jun 26. PMID: 24981835; PMCID: PMC4126889
  86. 86. Monda M, Sullo A, De Luca B. Lesions of the ventromedial hypothalamus reduce postingestional thermogenesis. Physiol Behav. 1997 May;61(5):687-691. DOI: 10.1016/s0031-9384(96)00520-3. PMID: 9145938
  87. 87. Monge-Roffarello B, Labbe SM, Lenglos C, Caron A, Lanfray D, Samson P, Richard D. The medial preoptic nucleus as a site of the thermogenic and metabolic actions of melanotan II in male rats. Am J Physiol Regul Integr Comp Physiol. 2014 Jul 15;307(2):R158-R166. DOI: 10.1152/ajpregu.00059.2014. PMID: 24808495
  88. 88. Rosenbaum M, Murphy EM, Heymsfield SB, Matthews DE, Leibel RL. Low dose leptin administration reverses effects of sustained weight-reduction on energy expenditure and circulating concentrations of thyroid hormones. J Clin Endocrinol Metab. 2002 May;87(5):2391-2394. DOI: 10.1210/jcem.87.5.8628. PMID: 11994393
  89. 89. usyniak DE, Zaretskaia MV, Zaretsky DV, DiMicco JA. Microinjection of muscimol into the dorsomedial hypothalamus suppresses MDMA-evoked sympathetic and behavioral responses. Brain Res. 2008 Aug 21;1226:116-23. DOI: 10.1016/j.brainres.2008.06.011. Epub 2008 Jun 14. PMID: 18586013; PMCID: PMC2600867
  90. 90. te Marie L, Miura GI, Marsh DJ, Yagaloff K, Palmiter RD. A metabolic defect promotes obesity in mice lacking melanocortin-4 receptors. Proc Natl Acad Sci U S A. 2000 Oct 24;97(22):12339-44. DOI: 10.1073/pnas.220409497. PMID: 11027312; PMCID: PMC17343
  91. 91. Tam CS, Lecoultre V, Ravussin E. Brown adipose tissue: mechanisms and potential therapeutic targets. Circulation. 2012 Jun 5;125(22):2782-2791. DOI: 10.1161/CIRCULATIONAHA.111.042929. PMID: 22665886.
  92. 92. van Marken Lichtenbelt W. Brown adipose tissue and the regulation of nonshivering thermogenesis. Curr Opin Clin Nutr Metab Care. 2012 Nov;15(6):547-552. DOI: 10.1097/MCO.0b013e3283599184. PMID: 23037904
  93. 93. Vaughan CH, Shrestha YB, Bartness TJ. Characterization of a novel melanocortin receptor-containing node in the SNS outflow circuitry to brown adipose tissue involved in thermogenesis. Brain Res. 2011 Sep 9;1411:17-27. DOI: 10.1016/j.brainres.2011.07.003. Epub 2011 Jul 13. PMID: 21802070; PMCID: PMC3426614
  94. 94. Welle S, Schwartz RG, Statt M. Reduced metabolic rate during beta-adrenergic blockade in humans. Metabolism. 1991 Jun;40(6):619-622. DOI: 10.1016/0026-0495(91)90053-y. PMID: 1650879
  95. 95. Zhou X, Gomez-Smith M, Qin Z, Duquette PM, Cardenas-Blanco A, Rai PS, Harper ME, Tsai EC, Anisman H, Chen HH. Ablation of LMO4 in glutamatergic neurons impairs leptin control of fat metabolism. Cell Mol Life Sci. 2012 Mar;69(5):819-28. DOI: 10.1007/s00018-011-0794-3. Epub 2011 Aug 27. PMID: 21874351; PMCID: PMC3276759
  96. 96. Jiang L, You J, Yu X, Gonzalez L, Yu Y, Wang Q, Yang G, Li W, Li C, Liu Y. Tyrosine-dependent and -independent actions of leptin receptor in control of energy balance and glucose homeostasis. Proc Natl Acad Sci U S A. 2008 Nov 25;105(47):18619-24. DOI: 10.1073/pnas.0804589105. Epub 2008 Nov 17. PMID: 19015522; PMCID: PMC2587560
  97. 97. Varela L, Horvath TL. Leptin and insulin pathways in POMC and AgRP neurons that modulate energy balance and glucose homeostasis. EMBO Rep. 2012 Dec;13(12):1079-86. DOI: 10.1038/embor.2012.174. Epub 2012 Nov 13. PMID: 23146889; PMCID: PMC3512417
  98. 98. Paz-Filho G, Mastronardi C, Franco CB, Wang KB, Wong ML, Licinio J. Leptin: molecular mechanisms, systemic pro-inflammatory effects, and clinical implications. Arq Bras Endocrinol Metabol. 2012 Dec;56(9):597-607. DOI: 10.1590/s0004-27302012000900001. PMID: 23329181
  99. 99. Carbone F, La Rocca C, Matarese G. Immunological functions of leptin and adiponectin. Biochimie. 2012 Oct;94(10):2082-2088. DOI: 10.1016/j.biochi.2012.05.018. Epub 2012 Jun 26. PMID: 22750129
  100. 100. Iikuni N, Lam QL, Lu L, Matarese G, La Cava A. Leptin and Inflammation. Curr Immunol Rev. 2008 May 1;4(2):70-79. DOI: 10.2174/157339508784325046. PMID: 20198122; PMCID: PMC2829991
  101. 101. Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med. 2012 Feb;33(1):35-45. DOI: 10.1016/j.mam.2011.10.012. Epub 2011 Oct 21. PMID: 22040697
  102. 102. Sharma V, Mustafa S, Patel N, Wambolt R, Allard MF, McNeill JH. Stimulation of cardiac fatty acid oxidation by leptin is mediated by a nitric oxide-p38 MAPK-dependent mechanism. Eur J Pharmacol. 2009 Sep 1;617(1-3):113-117. DOI: 10.1016/j.ejphar.2009.06.037. Epub 2009 Jun 30. PMID: 19573526
  103. 103. German JP, Thaler JP, Wisse BE, Oh-I S, Sarruf DA, Matsen ME, Fischer JD, Taborsky GJ Jr, Schwartz MW, Morton GJ. Leptin activates a novel CNS mechanism for insulin-independent normalization of severe diabetic hyperglycemia. Endocrinology. 2011 Feb;152(2):394-404. DOI: 10.1210/en.2010-0890. Epub 2010 Dec 15. PMID: 21159853; PMCID: PMC3037161
  104. 104. Hill JW, Williams KW, Ye C, Luo J, Balthasar N, Coppari R, Cowley MA, Cantley LC, Lowell BB, Elmquist JK. Acute effects of leptin require PI3K signaling in hypothalamic proopiomelanocortin neurons in mice. J Clin Invest. 2008 May;118(5):1796-805. DOI: 10.1172/JCI32964. PMID: 18382766; PMCID: PMC2276395
  105. 105. Agrawal S, Gollapudi S, Su H, Gupta S. Leptin activates human B cells to secrete TNF-α, IL-6, and IL-10 via JAK2/STAT3 and p38MAPK/ERK1/2 signaling pathway. J Clin Immunol. 2011 Jun;31(3):472-8. DOI: 10.1007/s10875-010-9507-1. Epub 2011 Jan 18. PMID: 21243519; PMCID: PMC3132280
  106. 106. Paz-Filho G, Mastronardi C, Franco CB, Wang KB, Wong ML, Licinio J. Leptin: molecular mechanisms, systemic pro-inflammatory effects, and clinical implications. Arq Bras Endocrinol Metabol. 2012 Dec;56(9):597-607. DOI: 10.1590/s0004-27302012000900001. PMID: 23329181
  107. 107. Burguera B, Couce ME, Long J, Lamsam J, Laakso K, Jensen MD, Parisi JE, Lloyd RV. The long form of the leptin receptor (OB-Rb) is widely expressed in the human brain. Neuroendocrinology. 2000 Mar;71(3):187-195. DOI: 10.1159/000054536. PMID: 10729790
  108. 108. Weigle DS, Duell PB, Connor WE, Steiner RA, Soules MR, Kuijper JL. Effect of fasting, refeeding, and dietary fat restriction on plasma leptin levels. J Clin Endocrinol Metab. 1997 Feb;82(2):561-565. DOI: 10.1210/jcem.82.2.3757. PMID: 9024254
  109. 109. Pelleymounter MA, Cullen MJ, Baker MB, Hecht R, Winters D, Boone T, Collins F. Effects of the obese gene product on body weight regulation in ob/ob mice. Science. 1995 Jul 28;269(5223):540-543. DOI: 10.1126/science.7624776. PMID: 7624776
  110. 110. Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK, Friedman JM. Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 1995 Jul 28;269(5223):543-546. DOI: 10.1126/science.7624777. PMID: 7624777
  111. 111. Satoh N, Ogawa Y, Katsuura G, Tsuji T, Masuzaki H, Hiraoka J, Okazaki T, Tamaki M, Hayase M, Yoshimasa Y, Nishi S, Hosoda K, Nakao K. Pathophysiological significance of the obese gene product, leptin, in ventromedial hypothalamus (VMH)-lesioned rats: evidence for loss of its satiety effect in VMH-lesioned rats. Endocrinology. 1997 Mar;138(3):947-954. DOI: 10.1210/endo.138.3.4989. PMID: 9048594
  112. 112. Roujeau C, Jockers R, Dam J. New pharmacological perspectives for the leptin receptor in the treatment of obesity. Front Endocrinol (Lausanne). 2014 Oct 13;5:167. DOI: 10.3389/fendo.2014.00167. PMID: 25352831; PMCID: PMC4195360
  113. 113. Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, Gojobori T, Isenovic ER. Leptin and Obesity: Role and Clinical Implication. Front Endocrinol (Lausanne). 2021 May 18;12:585887. DOI: 10.3389/fendo.2021.585887. PMID: 34084149; PMCID: PMC8167040
  114. 114. Gorden P, Gavrilova O. The clinical uses of leptin. Curr Opin Pharmacol. 2003 Dec;3(6):655-659. DOI: 10.1016/j.coph.2003.06.006. PMID: 14644019
  115. 115. Salvador J, Gomez-Ambrosi J, Frühbeck G. Perspectives in the therapeutic use of leptin. Expert Opin Pharmacother. 2001 Oct;2(10):1615-1622. DOI: 10.1517/14656566.2.10.1615. PMID: 11825304
  116. 116. Hukshorn CJ, Saris WH, Westerterp-Plantenga MS, Farid AR, Smith FJ, Campfield LA. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J Clin Endocrinol Metab. 2000 Nov;85(11):4003-4009. DOI: 10.1210/jcem.85.11.6955. PMID: 11095423
  117. 117. Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, Lubina JA, Patane J, Self B, Hunt P, McCamish M. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA. 1999 Oct 27;282(16):1568-1575. DOI: 10.1001/jama.282.16.1568. PMID: 10546697
  118. 118. Ring BD, Scully S, Davis CR, Baker MB, Cullen MJ, Pelleymounter MA, Danilenko DM. Systemically and topically administered leptin both accelerate wound healing in diabetic ob/ob mice. Endocrinology. 2000 Jan;141(1):446-449. DOI: 10.1210/endo.141.1.7373. PMID: 10614668
  119. 119. Yamauchi T, Waki H, Kamon J, Murakami K, Motojima K, Komeda K, Miki H, Kubota N, Terauchi Y, Tsuchida A, Tsuboyama-Kasaoka N, Yamauchi N, Ide T, Hori W, Kato S, Fukayama M, Akanuma Y, Ezaki O, Itai A, Nagai R, Kimura S, Tobe K, Kagechika H, Shudo K, Kadowaki T. Inhibition of RXR and PPARgamma ameliorates diet-induced obesity and type 2 diabetes. J Clin Invest. 2001 Oct;108(7):1001-13. DOI: 10.1172/JCI12864. PMID: 11581301; PMCID: PMC200951
  120. 120. Cohen P, Miyazaki M, Socci ND, Hagge-Greenberg A, Liedtke W, Soukas AA, Sharma R, Hudgins LC, Ntambi JM, Friedman JM. Role for stearoyl-CoA desaturase-1 in leptin-mediated weight loss. Science. 2002 Jul 12;297(5579):240-243. DOI: 10.1126/science.1071527. PMID: 12114623
  121. 121. Lindsay RS, Funahashi T, Hanson RL, Matsuzawa Y, Tanaka S, Tataranni PA, Knowler WC, Krakoff J. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet. 2002 Jul 6;360(9326):57-58. DOI: 10.1016/S0140-6736(02)09335-2. PMID: 12114044
  122. 122. Spranger J, Kroke A, Möhlig M, Bergmann MM, Ristow M, Boeing H, Pfeiffer AF. Adiponectin and protection against type 2 diabetes mellitus. Lancet. 2003 Jan 18;361(9353):226-8. DOI: 10.1016/S0140-6736(03)12255-6. Erratum in: Lancet. 2002 Mar 22;361(9362):1060. PMID: 12547549
  123. 123. Sanna V, Di Giacomo A, La Cava A, Lechler RI, Fontana S, Zappacosta S, Matarese G. Leptin surge precedes onset of autoimmune encephalomyelitis and correlates with development of pathogenic T cell responses. J Clin Invest. 2003 Jan;111(2):241-50. DOI: 10.1172/JCI16721. PMID: 12531880; PMCID: PMC151876
  124. 124. Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, O'Rahilly S. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002 Oct;110(8):1093-103. DOI: 10.1172/JCI15693. PMID: 12393845; PMCID: PMC150795
  125. 125. Moran S, Young J, Ruiz E, Sebring N, Premkumar A, Depaoli A, Oral E, Skarulis M, Gorden P: Changes in body composition in patients with lipodystrophic diabetes after leptin replacement therapy. Diabetes 2003, 52:A76
  126. 126. Frühbeck G. Intracellular signalling pathways activated by leptin. Biochem J. 2006 Jan 1;393(Pt 1):7-20. DOI: 10.1042/BJ20051578. PMID: 16336196; PMCID: PMC1383660
  127. 127. Hileman SM, Tornøe J, Flier JS, Bjørbaek C. Transcellular transport of leptin by the short leptin receptor isoform ObRa in Madin-Darby Canine Kidney cells. Endocrinology. 2000 Jun;141(6):1955-1961. DOI: 10.1210/endo.141.6.7450. PMID: 10830277
  128. 128. Arvaniti K, Huang Q, Richard D. Effects of leptin and corticosterone on the expression of corticotropin-releasing hormone, agouti-related protein, and proopiomelanocortin in the brain of ob/ob mouse. Neuroendocrinology. 2001 Apr;73(4):227-236. DOI: 10.1159/000054639. PMID: 11340336
  129. 129. Pandit R, Beerens S, Adan RAH. Role of leptin in energy expenditure: the hypothalamic perspective. Am J Physiol Regul Integr Comp Physiol. 2017 Jun 1;312(6):R938-R947. DOI: 10.1152/ajpregu.00045.2016. Epub 2017 Mar 29. PMID: 28356295
  130. 130. hao S, Zhu Y, Schultz RD, Li N, He Z, Zhang Z, Caron A, Zhu Q, Sun K, Xiong W, Deng H, Sun J, Deng Y, Kim M, Lee CE, Gordillo R, Liu T, Odle AK, Childs GV, Zhang N, Kusminski CM, Elmquist JK, Williams KW, An Z, Scherer PE. Partial Leptin Reduction as an Insulin Sensitization and Weight Loss Strategy. Cell Metab. 2019 Oct 1;30(4):706-719.e6. DOI: 10.1016/j.cmet.2019.08.005. Epub 2019 Sep 5. PMID: 31495688; PMCID: PMC6774814

Written By

Reji Manjunathan, Dharanibalan Kasiviswanathan and Selvaraj Jayaraman

Submitted: 12 September 2021 Reviewed: 21 September 2021 Published: 09 October 2021