1
|
Yupanqui-Lozno H, Huertas-Quintero JA, Yupanqui-Velazco ME, Salinas-Osornio RA, Restrepo CM, Gonzalez A, Nava-Gonzalez EJ, Celis-Regalado LG, Neri Morales C, Hernandez-Escalante VM, Licinio J, Laviada-Molina HA, Rodriguez-Ayala E, Arango C, Bastarrachea RA. One-year metreleptin in Colombian sisters with congenital leptin deficiency. Adipocyte 2025; 14:2508188. [PMID: 40415699 PMCID: PMC12118419 DOI: 10.1080/21623945.2025.2508188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 02/27/2025] [Accepted: 05/02/2025] [Indexed: 05/27/2025] Open
Abstract
We discovered two adult sisters in Colombia, lineally consanguineous, with severe obesity and undetectable serum leptin levels despite markedly elevated body fat. Their clinical profile included childhood-onset extreme weight gain, intense hunger, hyperphagia, hypogonadotropic hypogonadism, and family history of obesity. Direct sequencing of the LEP gene revealed a novel homozygous missense mutation in exon 3 (c.350G>T [p.C117F]). The presence of this mutation, undetectable leptin, and severe obesity confirmed a diagnosis of monogenic leptin deficiency. Here we describe the clinical outcomes of a 12-month treatment with recombinant human leptin (metreleptin). Metabolic and endocrine assessments were conducted before and after therapy. Metreleptin therapy significantly reduced BMI: from 59 to 38 kg/m2 (OBX1, age 27) and 60 to 48 kg/m2 (OBX2, age 24). Total body fat mass decreased, serum lipids normalized, and insulin sensitivity improved. Hypogonadotropic hypogonadism reversed, and menstruation resumed. Thus, metreleptin reversed the major metabolic and endocrine abnormalities associated with leptin deficiency in these sisters. Limitations include the small sample size, absence of a control group, and lack of anti-metreleptin antibody measurements. Nevertheless, our findings support that leptin replacement with metreleptin is currently the only effective hormonal treatment for this monogenic form of human obesity.
Collapse
Affiliation(s)
| | - Jancy Andrea Huertas-Quintero
- Clinical Research Department, Dexa Diab IPS, Bogotá, Colombia
- Hospital Universitario Mayor, Universidad del Rosario, Bogotá, Colombia
| | | | - Rocío A. Salinas-Osornio
- Centro de Investigación y Desarrollo Científico, Ciencias de la Salud, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | - Carlos M. Restrepo
- GeniURos, CIGGUR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | | | - Edna J. Nava-Gonzalez
- Facultad de Salud Pública y Nutrición (Faspyn), Universidad Autónoma de Nuevo León, Monterrey, México
| | | | | | | | - Julio Licinio
- Department of Genetics, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | | | - Carlos Arango
- Hospital Universitario Mayor, Universidad del Rosario, Bogotá, Colombia
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
2
|
Zhang W, Peng J, Wang N, Shi Z, Wu J, Tong D. Expression of leptin and its long-form receptor in the porcine corpus luteum during pregnancy and the protective role of leptin in corpus luteum function invitro. Theriogenology 2025; 242:117402. [PMID: 40250250 DOI: 10.1016/j.theriogenology.2025.117402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
Although the hormone leptin and its long-form receptor (LEPR) have been identified in the porcine ovary, their direct roles in mediating corpus luteum (CL) function in pregnant sows have not been clearly defined. In this study, we investigated leptin and LEPR expression in the porcine CL during pregnancy and evaluated the effect of the hormone on CL function in vitro. Leptin and LEPR were continuously expressed in the CL throughout pregnancy, and their expression patterns were generally similar, with higher levels in the early and middle stages of pregnancy and lower levels in the late stage. Immunohistochemical analysis revealed that both leptin and LEPR were predominantly localized in the luteal cells, with strong immunostaining observed in the early and middle stages CL and weak immunostaining in the late stage. Leptin 1-100 nM concentrations significantly increased the viability of cultured luteal cells (P < 0.05) and promoted their release of progesterone (P4) in a dose-dependent manner. Furthermore, leptin attenuated the prostaglandin F2α (PGF2α)-induced decreases in cell viability and P4 release. Additionally, leptin was found to have stimulatory effects on luteinizing hormone (LH) and prostaglandin E2 (PGE2) release but an inhibitory effect on PGF2α secretion. In conclusion, the presence of leptin and LEPR in the CL and the stimulatory effects of the hormone on the viability of luteal cells and their release of P4, LH, and PGE2 suggest that leptin has a positive regulatory effect on CL function in pregnant sows.
Collapse
Affiliation(s)
- Wenlong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Jiang Peng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Ning Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zulian Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Jiaman Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Dewen Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
3
|
Tokdemir M, Erbak E, Tunçez FT, Elmali F, Yilmaz HE. Evaluation of leptin, insulin, orexin, neuropeptide y (NPY) levels in postmortem CSF samples in suicide deaths. J Affect Disord 2025; 381:303-309. [PMID: 40187429 DOI: 10.1016/j.jad.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Suicide remains a significant global public health issue. According to the World Health Organization (WHO), suicide was the third leading cause of mortality among individuals aged 15-29 in 2021, with a total of approximately 726,000 cases reported annually. The etiology of suicide is complex, involving a combination of biological, genetic, and environmental factors, as well as family history, gender, age, personality traits, cultural background, geographic location, medical conditions, mental illnesses, addictions, and psychosocial stressors. Dysregulation of the Hypothalamic Pituitary Adrenal (HPA) axis and the effects of chronic stress play significant roles in the pathophysiology of mood disorders and suicidal behavior. OBJECTIVE This study aimed to investigate the levels of Neuropeptide Y (NPY), Orexin, Leptin, and Insulin in cerebrospinal fluid (CSF) samples of individuals who died by suicide compared to those who died from non-suicidal causes. METHOD The study examined 35 cases of suicide by hanging and 35 cases of non-suicidal deaths unrelated to head trauma. Levels of NPY, Orexin, Leptin, and Insulin in CSF samples collected during toxicological examinations were compared between suicide and control groups. RESULTS NPY levels were significantly higher in the suicide group than in the control group (p < 0.001). No statistically significant differences were found in Orexin (p = 0.194), Insulin (p = 0.892), or Leptin (p = 0.445) levels between the groups. CONCLUSIONS While no definitive biomarkers for diagnosing or predicting suicidal behavior exist, this panel of biomarkers could provide valuable insights for developing targeted treatments to manage patients at risk.
Collapse
Affiliation(s)
- Mehmet Tokdemir
- Izmir Katip Celebi University School of Medicine, Department of Forensic Medicine, Izmir, Turkey; Council of Forensic Medicine Chairmanship of Group, Izmir, Turkey.
| | - Esra Erbak
- Izmir Katip Celebi University School of Medicine, Department of Forensic Medicine, Izmir, Turkey
| | - Ferhat Turgut Tunçez
- Izmir Katip Celebi University School of Medicine, Department of Forensic Medicine, Izmir, Turkey
| | - Ferhan Elmali
- Izmir Katip Celebi University School of Medicine, Department of Biostatistics, Izmir, Turkey
| | - Huriye Erbak Yilmaz
- Izmir Katip Celebi University School of Medicine, Department of Biochemistry, Izmir, Turkey; Izmir Biomedicine and Genome Center, Izmir, Turkey
| |
Collapse
|
4
|
Teng TQ, Wang MM, Mo DG, Xie YY, Chen R, Xu JC, Liu J, Yu HC. Synergistic effects of a body shape index and depression on mortality in individuals with low sexual frequency. J Affect Disord 2025; 380:104-112. [PMID: 40122262 DOI: 10.1016/j.jad.2025.03.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Individuals with low sexual frequency often experience comorbidities that exacerbate mortality. This article evaluates the predictive value of five body fat anthropometric indicators for all-cause mortality and explores the interaction between obesity and depression in mortality among young and middle-aged individuals with sexual frequency <12 times per year. METHODS This study included participants with a sexual frequency of <12 times per year from the 2015-2016 National Health and Nutrition Examination Survey (NHANES). We assessed the impact of anthropometric indicators and depression on mortality, as well as their synergistic interactions, and further developed an accessible predictive survival model. RESULTS A total of 4978 participants aged 20-59 were included, with 215 deaths (4.3 %) over 15 years of follow-up. A Body Shape Index (ABSI) showed the strongest association with all-cause mortality, with an AUC of 0.67. Participants with ABSI ≥0.082 had a significantly higher risk of death (HR: 1.87, 95%CI: 1.31-2.68), as did those with depression (HR: 1.86, 95%CI: 1.19-2.92). Interaction analysis revealed a synergistic effect between depression and ABSI, increasing death risk by 293 % when both were present. Significant survival differences were observed between men and women with these risk factors, with median survival rates of 76.3 % and 90.8 %, respectively. The model based on ABSI and depression provided valuable mortality predictions, with AUC of 0.78, 0.77, and 0.77 for 3-year, 5-year, and 10-year survival. CONCLUSION ABSI and depression are associated with all-cause mortality in individuals with low sexual frequency, potentially creating a synergistic effect on mortality risk.
Collapse
Affiliation(s)
- Tian-Qi Teng
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao, Shandong 266000, China
| | - Meng-Meng Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao, Shandong 266000, China
| | - De-Gang Mo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yan-You Xie
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Rui Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Jia-Chao Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Jing Liu
- Department of Neurology, Xuzhou New Health Geriatric Hospital, Xuzhou, Jiangsu 221000, China.
| | - Hai-Chu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
5
|
Qiao C, Qin X, Song Y, Guan R, Li B, Zuo Y, Wei W, Han T, Jiang W. Association of childhood emotional neglect, circulating protein biomarkers, with gastrointestinal disorders among UK biobank participants. J Affect Disord 2025; 380:317-330. [PMID: 40120955 DOI: 10.1016/j.jad.2025.03.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE To explore the association between CEN and GIDs, and elucidated the potential role of circulating protein biomarkers. PATIENTS AND METHODS The study utilized UK Biobank data from 156,686 participants, with data collection occurring between March 13, 2006 and October 1, 2010. Participants with GIDs at baseline were excluded from further analysis. CEN data were obtained from the baseline assessments. Differential protein analyses were conducted using OLINK data. GIDs and their subclasses were identified through electronic health records. Cox proportional hazards regression models were employed to assess the association between CEN and the risk of GIDs, along with sensitivity and multidimensional stratification analyses. Additionally, mediation analysis was performed to explore the role of differential protein biomarkers. RESULTS The results indicated that the mild CEN (CENmild) group was associated with a significantly lower risk of various GIDs than the severe CEN (CENsevere) group, including overall GIDs (HR = 0.78,95%CI:0.74-0.81) and peptic ulcers (HR = 0.37,95%CI:0.20-0.68). OLINK differential analysis revealed that APOF expression was significantly higher in the CENmild group compared to the CENsevere group (PAPOF = 7.09E-08,FC = 0.048), whereas other differential protein expression (PBPIFB2 = 8.93E-06,FC = -0.122;PFABP4 = 3.19E-06,FC = -0.101;PGGH = 4.58E-07,FC = -0.054;PLEP = 5.39E-08,FC = -0.195) was significantly lower in the CENsevere group. Cox regression analysis showed that higher APOF expression was associated with a reduced risk of multiple GIDs, while the expression of other differential proteins increased the risk of corresponding GIDs. Mediation analysis indicated that these proteins mediated 0.5 % to 6.7 % of the CEN-GIDs association. CONCLUSION In this cohort study, CEN was significantly associated with a higher risk of GIDs in the adulthood, and circulating protein biomarkers partially mediated the associations.
Collapse
Affiliation(s)
- Conghui Qiao
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Xiaowen Qin
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Yuqing Song
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Ruijie Guan
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Bai Li
- Department of Biology, University of Ottawa, 30 Marie-Curie Private, Gendron Hall, Ottawa, ON K1N 9B4, Canada
| | - Yingdong Zuo
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China.
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China.
| | - Wenbo Jiang
- Department of Nutrition and Food Hygiene, the National Key Discipline, School of Public Health, Harbin Medical University, Harbin, PR China; Department of Biology, University of Ottawa, 30 Marie-Curie Private, Gendron Hall, Ottawa, ON K1N 9B4, Canada.
| |
Collapse
|
6
|
Arfuso F, Arrigo F, Lopreiato V, Rizzo M, Liotta L, Giannetto C, Piccione G. Changes in plasma UCP-1, leptin, lipids, and pro-inflammatory interleukins in calves from birth to weaning. Res Vet Sci 2025; 189:105627. [PMID: 40154030 DOI: 10.1016/j.rvsc.2025.105627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/24/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
The neonatal period is crucial for calf development, particularly for immune acquisition through colostrum intake, this study aimed to assess the energy metabolism and inflammatory response. Ten Italian Simmental calves were monitored from birth to 60 days of age, with blood samples taken at birth (0 d) to weaning. Plasma concentrations of UCP-1, leptin, TNF-α, IL-1β, IL-6, lipids, triglycerides, and total cholesterol were measured. Results showed significant dynamic changes (P < 0.05) in all parameters and showing an increasing trend from birth to the 60 d of age in investigated calves. The plasma leptin levels were positively correlated with the concentration of UCP-1 (r = 0.37, P = 0.0003), total lipids (r = 0.47, P < 0.0001), triglycerides (r = 0.53, P < 0.0001), total cholesterol (r = 0.38, P = 0.0002), and negatively correlated with TNF-α (r = -0.24, P 0.02). UCP-1 was positively correlated with the levels of total lipids (r = 0.31, P = 0.003), triglycerides (r = 0.29, P = 0.005), and IL-1β (r = 0.29, P = 0.005) in calves throughout the monitoring period. IL-6 values positively correlated with total lipids (r = 0.36; P = 0.0004), triglycerides (r = 0.37; P = 0.0003), and total cholesterol (r = 0.21; P = 0.04) in calves throughout the monitoring period. These findings suggest that lipid metabolism and inflammatory responses undergo significant changes as calves adapt to the neonatal phase and transition to solid food, with nutritional shifts playing a key role in metabolic and immune system development.
Collapse
Affiliation(s)
- Francesca Arfuso
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Federica Arrigo
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Vincenzo Lopreiato
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Maria Rizzo
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Luigi Liotta
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Claudia Giannetto
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| | - Giuseppe Piccione
- Department of Veterinary Sciences, Viale Giovanni Palatucci snc, University of Messina, 98168 Messina, Italy.
| |
Collapse
|
7
|
Cheong ASQ, Suvan JE. Considerations in the treatment of individuals with obesity and periodontitis. Clin Obes 2025; 15:e70002. [PMID: 40135427 PMCID: PMC12096047 DOI: 10.1111/cob.70002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 12/28/2024] [Accepted: 02/05/2025] [Indexed: 03/27/2025]
Abstract
Two common non-communicable diseases, obesity and periodontitis, are responsible for and affected by systemic inflammation, sharing common risk factors and mechanistic pathways. Periodontitis is an irreversible immune-mediated inflammatory disease of hard and soft tissue supporting teeth. If left untreated, periodontitis can lead to tooth loss, affecting food choices and healthy eating, therefore affecting overall health. Obesity is an independent predictive factor for worsened periodontal inflammation, increased onset, progression, severity, and recurrence of infection, as well as delayed wound healing. Thus, managing obesity and associated metabolic dysfunctions may improve periodontal therapy outcomes. The chronic inflammatory state of obesity impairs immune regulation exacerbating the inflammatory gingival tissue destruction of periodontitis, which can also systemically contribute to inflammatory mediators. Furthermore, bariatric surgeons and dietitians should educate patients with obesity regarding the risk of elevated caries, xerostomia, and periodontitis risk from acid reflux and frequent food intake. Non-dental healthcare professionals should recognise periodontal disease signs to prompt dental referral when warranted. Asking patients about recent dental visits promotes patient involvement in cross-discipline dialogue to enhance patient care coordination between medicine and dentistry. This article discusses the association between these two diseases, the challenges of achieving optimal periodontal treatment outcomes, and the clinical strategies to enhance holistic care. It also explores oral health considerations in dietary and surgical interventions in the treatment of obesity.
Collapse
Affiliation(s)
| | - Jean E. Suvan
- Oral Sciences, University of Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
8
|
Nehus E, Mitsnefes M. Kidney consequences of obesity. Pediatr Nephrol 2025; 40:1879-1893. [PMID: 39680134 DOI: 10.1007/s00467-024-06623-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
Herein, we review the devastating consequences of the worldwide obesity epidemic on kidney health and outcomes. We submit that the obesity epidemic is the most pressing public health crisis facing the nephrology community today. A historical approach has been undertaken, wherein major breakthroughs in the recognition and understanding of obesity-related kidney disease (ORKD) are highlighted. We begin with a description of the worldwide obesity epidemic followed by an account of the discovery and characterization of ORKD. A detailed summary of the pathophysiology of ORKD disease is presented, wherein we set forth the following two propositions: first, ORKD is due to a maladaptive response to caloric surplus; and second, this maladaptive response causes kidney damage via hemodynamic (hyperfiltration), hormonal (adipokine dysregulation), and lipotoxic pathways. Each of these pathways is described, with particular emphasis on the relatively recent discovery that the final stage of cellular injury in ORKD is mitochondrial oxidative damage. The prevention and treatment of ORKD are then discussed, including environmental, behavioral, pharmacologic, and surgical options. Finally, we conclude with suggestions for future research to improve early recognition and treatment of ORKD.
Collapse
Affiliation(s)
- Edward Nehus
- Department of Pediatrics, West Virginia University School of Medicine Charleston Campus, Charleston, WV, 25314, USA.
- Institute for Academic Medicine, Charleston Area Medical Center, Charleston, WV, USA.
| | - Mark Mitsnefes
- Division of Nephrology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
9
|
Nasr M, Youssef M, Alghamdi AAA, Alghamdi AH, Khormi MA, Aborasain AM, Emeish WFA, Elkamel AA. Effects of ammonia exposure on the expression of IL-1β, CRH, and lep-a1 genes in common carp (Cyprinus carpio). BMC Vet Res 2025; 21:383. [PMID: 40437528 PMCID: PMC12117808 DOI: 10.1186/s12917-025-04749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/10/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Common carp is one of the most economically important freshwater fish species globally. Ammonia exposure, a frequent challenge in aquaculture, can lead to significant economic losses. This study investigated the impact of un-ionized ammonia (UIA) exposure on the expression profiles of three key genes in common carp fry: interleukin-1 beta (IL-1β), corticotropin-releasing hormone (CRH), and leptin a1 (Lep-a1). These genes are crucial indicators of immune response, stress regulation, and appetite control, respectively. Fish were exposed to 0.7 mg/L of UIA, and gene expression was analysed in liver and gill tissues at five time points (12 h, 2-, 4-, 7-, and 14-days of exposure) using quantitative real-time PCR (RT-qPCR). RESULTS Results demonstrated that expression levels of all three genes were significantly affected by exposure time and tissue type. IL-1β, CRH, and Lep-a1 were upregulated in both liver and gill tissues, with the liver consistently showing higher expression levels. Notably, significant positive correlations were observed between each pair of the three genes studied, suggesting a coordinated physiological response to ammonia stress. The liver emerged as a key organ in orchestrating the long-term adaptive response, while the gills exhibited a more acute, transient reaction. CONCLUSIONS This study provides valuable insights into the molecular mechanisms underlying the physiological response of common carp to ammonia toxicity. The findings highlight the complex interplay between immune, stress, and metabolic pathways in coping with ammonia exposure. A deep understanding of these mechanisms could lead to improved management strategies in aquaculture and the development of potential biomarkers for assessing stress responses in fish populations.
Collapse
Affiliation(s)
- Mahmoud Nasr
- Department of Fish Diseases, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Mohammed Youssef
- Department of Animal Physiology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Abdullah A A Alghamdi
- Department of Biology, Faculty of Science, Al-Baha University, Al-Baha, Saudi Arabia
| | - Ali H Alghamdi
- Department of Biology, Faculty of Science, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohsen A Khormi
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan, 45142, Saudi Arabia
| | - Ali M Aborasain
- Department of Biology, College of Science, Jazan University, P.O. Box. 114, Jazan, 45142, Saudi Arabia
| | - Walaa F A Emeish
- Department of Fish Diseases, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Ahmad A Elkamel
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
10
|
Salyer LG, Wang Y, Ma X, Foryst-Ludwig A, Kintscher U, Chennappan S, Kontaridis MI, McKinsey TA. Modulating the Secretome of Fat to Treat Heart Failure. Circ Res 2025; 136:1363-1381. [PMID: 40403114 DOI: 10.1161/circresaha.125.325593] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/24/2025]
Abstract
Heart failure afflicts >6 million individuals in the United States alone and is associated with significant mortality (≈40% within 5 years of diagnosis) and cost (estimated to exceed $70 billion in the United States by 2030). Obesity is a major risk factor for the development of heart failure. The contribution of excess adipose tissue to heart failure pathogenesis is multifactorial. For example, adipose tissue-driven inflammation contributes to the development of other cardiometabolic comorbidities, such as hypertension, leading to left ventricular pressure overload and adverse remodeling of the heart. Adipose tissue also functions as an endocrine organ, and altered secretion of proteins, lipid mediators, metabolites, and small extracellular vesicles (collectively referred to as the secretome) from dysfunctional fat can lead to cardiac inflammation and oxidative stress, which drive changes in structure and function of the heart. In this review, we begin with an overview of current therapies for obesity and what is known about how they influence the heart. Then we focus on mechanisms by which fat communicates with the heart via secreted factors and highlight druggable nodes in this circuit that could be exploited to develop next-generation therapies for heart failure.
Collapse
Affiliation(s)
- Lorien G Salyer
- Division of Cardiology, Department of Medicine (L.G.S., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora, CO
- Consortium for Fibrosis Research & Translation (L.G.S., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL (Y.W.)
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA (X.M.)
| | - Anna Foryst-Ludwig
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany (A.F.-L., U.K.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (A.F.-L., U.K.)
| | - Ulrich Kintscher
- Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Germany (A.F.-L., U.K.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (A.F.-L., U.K.)
| | - Saravanakkumar Chennappan
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY (S.C., M.I.K.)
| | - Maria I Kontaridis
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY (S.C., M.I.K.)
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA (M.I.K.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA (M.I.K.)
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine (L.G.S., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora, CO
- Consortium for Fibrosis Research & Translation (L.G.S., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
11
|
Sun X, Xin C, Yao J, Wang H. Relationship Between Leptin and Heart Failure: A Meta-Analysis. Glob Heart 2025; 20:44. [PMID: 40417460 PMCID: PMC12101114 DOI: 10.5334/gh.1434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Background Heart failure (HF) is a diverse and potentially fatal condition affecting more than 60 million people worldwide. Previous studies have identified a close relationship between leptin levels and HF, and that leptin levels in patients with HF are higher than those in healthy individuals. However, some studies have reported inconsistent results. Therefore, the association between leptin levels and HF remains controversial. Methods A literature search was conducted on the Web of Science, Wiley Online Library, Embase, and PubMed databases. The title or abstract search term 'leptin' was used in combination with 'heart failure' and 'HF'. Meta-analysis results were reported as standardized mean differences (SMD) with corresponding 95% confidence intervals (CI). Results Eighteen studies comprising 1149 patients with HF and 622 healthy controls were included in the meta-analysis. Leptin levels in patients with HF were significantly higher than those in healthy individuals (SMD, 0.54; 95% CI [0.15, 0.93]). Conclusions To our knowledge, this systematic review is the first to evaluate the relationship between leptin and HF. Determining the role of leptin in HF will significantly contribute to its diagnosis and treatment.
Collapse
Affiliation(s)
- Xin Sun
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, CN
| | - Caihong Xin
- Department of Endocrinology and Metabolism, The Fourth People’s Hospital of Soochow University, Shenyang, CN
| | - Jiayi Yao
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou, CN
| | - Hongli Wang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, CN
| |
Collapse
|
12
|
Ziegler KA, Engelhardt S, Carnevale D, McAlpine CS, Guzik TJ, Dimmeler S, Swirski FK. Neural Mechanisms in Cardiovascular Health and Disease. Circ Res 2025; 136:1233-1261. [PMID: 40403111 DOI: 10.1161/circresaha.125.325580] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/22/2025] [Accepted: 04/22/2025] [Indexed: 05/24/2025]
Abstract
Although the neurocardiac axis is central to cardiovascular homeostasis, its dysregulation drives heart failure and cardiometabolic diseases. This review examines the bidirectional interplay between the autonomic nervous system and the heart, highlighting the role of this interplay in disease progression and its therapeutic potential. The autonomic nervous system modulates cardiac function and vascular tone through its sympathetic and parasympathetic branches. However, in heart failure, chronic sympathetic overdrive and parasympathetic withdrawal exacerbate myocardial remodeling and metabolic dysfunction, both of which are exacerbated by cardiometabolic conditions such as obesity and diabetes. These conditions are increasingly recognized to impair neurocardiac regulation, thereby promoting inflammation and adverse outcomes. An important emerging area concerns neuroimmune control, in which the brain orchestrates systemic inflammation through circuits involving the bone marrow, spleen, and other organs, thereby amplifying cardiovascular damage. This neuroimmune axis integrates peripheral signals to influence immune responses that contribute to disease progression. Lifestyle factors, such as stress, sleep, exercise, and diet, affect autonomic and immune balance and, thus, cardiovascular disease. Therapeutically, targeting neurocardiac and neuroimmune pathways pharmacologically or via neuromodulation (eg, vagal or splenic nerve stimulation) offers promise although the clinical translation of the latter remains challenging. In this review, we synthesize preclinical and clinical data to highlight the neurocardiac axis as a critical nexus in heart failure and cardiometabolic disease. Harnessing neuroimmune and neurocardiac interactions may inform precision approaches to reduce the burden of these conditions.
Collapse
Affiliation(s)
- Karin A Ziegler
- Institute of Pharmacology and Toxicology, School of Medicine and Health, Technical University of Munich, Germany (K.A.Z., S.E.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (K.A.Z., S.E.)
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, School of Medicine and Health, Technical University of Munich, Germany (K.A.Z., S.E.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany (K.A.Z., S.E.)
| | - Daniela Carnevale
- Faculty of Pharmacy and Medicine, Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (D.C.)
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy (D.C.)
| | - Cameron S McAlpine
- Cardiovascular Research Institute, The Friedman Brain Institute, and Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.S.M., F.K.S.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, The University of Edinburgh, United Kingdom (T.J.G.)
- Department of Internal Medicine (T.J.G.), Jagiellonian University Medical College, Kraków, Poland
- Center for Medical Genomics OMICRON (T.J.G.), Jagiellonian University Medical College, Kraków, Poland
| | - Stefanie Dimmeler
- Goethe University Frankfurt, Institute for Cardiovascular Regeneration, Germany (S.D.)
- German Centre for Cardiovascular Research (DZHK), Frankfurt am Main, Germany (S.D.)
- Cardiopulmonary Institute, Goethe University Frankfurt am Main, Germany (S.D.)
| | - Filip K Swirski
- Cardiovascular Research Institute, The Friedman Brain Institute, and Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY (C.S.M., F.K.S.)
| |
Collapse
|
13
|
Sabaratnam R. The endocrine effects of leptin on energy metabolism. Nat Rev Endocrinol 2025:10.1038/s41574-025-01135-7. [PMID: 40404935 DOI: 10.1038/s41574-025-01135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research & Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
14
|
Rodrigues FM, Majeres LE, Dilger AC, McCann JC, Cassady CJ, Shike DW, Beever JE. Characterizing differences in the muscle transcriptome between cattle with alternative LCORL-NCAPG haplotypes. BMC Genomics 2025; 26:479. [PMID: 40369436 PMCID: PMC12076881 DOI: 10.1186/s12864-025-11665-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND The LCORL-NCAPG locus is a major quantitative trait locus (QTL) on bovine chromosome 6 (BTA6) that influences growth and carcass composition in cattle. To further understand the molecular mechanism responsible for the phenotypic changes associated with this locus, twenty-four Charolais-sired calves were selected for muscle transcriptome analysis based on alternative homozygous LCORL-NCAPG haplotypes (i.e., 12 "QQ" and 12 "qq", where "Q" is a haplotype harboring variation associated with increased growth). At 300 days of age, a biopsy of the longissimus dorsi muscle was collected from each animal for RNA sequencing. RESULTS Gene expression analysis identified 733 genes as differentially expressed between QQ and qq animals (q-value < 0.05). Notably, LCORL and genes known to be important regulators of growth such as IGF2 were upregulated in QQ individuals, while genes associated with adiposity such as FASN and LEP were downregulated, reflecting the increase in lean growth associated with this locus. Gene set enrichment analysis demonstrated QQ individuals had downregulation of pathways associated with adipogenesis, alongside upregulation of transcripts for cellular machinery essential for protein synthesis and energy metabolism, particularly ribosomal and mitochondrial components. CONCLUSIONS The differences in the muscle transcriptome between QQ and qq animals imply that muscle hypertrophy may be metabolically favored over accumulation of fat in animals with the QQ haplotype. Our findings also suggest this haplotype could be linked to a difference in LCORL expression that potentially influences the downstream transcriptional effects observed, though further research will be needed to confirm the molecular mechanisms underlying the associated changes in phenotype.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Division of Biological and Biomedical Sciences, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Leif E Majeres
- Department of Animal Science and Large Animal Clinical Sciences, University of Tennessee Institute of Agriculture, Knoxville, TN, USA
| | - Anna C Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joshua C McCann
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Christopher J Cassady
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Dan W Shike
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jonathan E Beever
- Department of Animal Science and Large Animal Clinical Sciences, University of Tennessee Institute of Agriculture, Knoxville, TN, USA.
| |
Collapse
|
15
|
Morath V, Maurer S, Feuchtinger A, Walser R, Schlapschy M, Bolze F, Metzler T, Bruder J, Steiger K, Walch A, Klingenspor M, Skerra A. Long-Acting Human PASylated Leptin Reaches the Murine Central Nervous System and Offers Potential for Optimized Replacement Therapy. Mol Pharm 2025. [PMID: 40335095 DOI: 10.1021/acs.molpharmaceut.4c01503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Despite the multifaceted role of leptin for energy homeostasis and its broad therapeutic potential, the FDA/EMA-approved metreleptin constitutes the only leptin drug to date. To translate the promising results from previous studies on murine PASylated leptin with improved solubility and extended plasma half-life using PASylation technology─a biological alternative to PEGylation─we have developed a second-generation human leptin drug candidate and tested it rigorously in vitro and in vivo. To this end, the exposed hydrophobic Trp residue at position 100 in human leptin was replaced by Gln, which, together with the genetic fusion with a 600-residue PAS polypeptide, yielded a protein with high solubility, folding stability and receptor-stimulatory activity. In a pharmacokinetic (PK) study with wild-type mice, this modified human leptin showed an extended plasma half-life of 18.8 ± 3.6 h after subcutaneous (s.c.) injection. Furthermore, leptin-deficient mice were dosed s.c. with the modified human leptin carrying two different PAS fusion tags, PAS#1 or P/A#1, each comprising 600 residues. After only four doses, the disease phenotype, including morbid adiposity, hyperphagia, and hepatic steatosis, was completely reversed by both PASylated leptin versions, but not by the non-PASylated leptin if administered at the same dose. To assess its tissue distribution, P/A(200)-huLeptinW100Q was doubly labeled with two fluorescent dyes, which were specifically attached to the leptin and the PAS moiety, respectively. Analysis of relevant mouse organs by light sheet fluorescence microscopy after clearance revealed colocalized signals in the kidney and liver, thus indicating general stability of the PAS-leptin fusion protein in vivo. However, discrete signals were observed in the hypothalamic region, only with leptin detectable in the choroid plexus, which implies cleavage of the PAS tag during transcytosis across the physiological barriers. This study should pave the way toward a second-generation leptin drug enabling prolonged dosing intervals.
Collapse
Affiliation(s)
- Volker Morath
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
- Department of Nuclear Medicine, School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
| | - Stefanie Maurer
- Chair for Molecular Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan 85354, Germany
- EKFZ─Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich 81675, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Rebecca Walser
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Martin Schlapschy
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| | - Florian Bolze
- Chair for Molecular Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan 85354, Germany
- EKFZ─Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich 81675, Germany
| | - Thomas Metzler
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
| | - Johanna Bruder
- Chair for Molecular Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan 85354, Germany
- EKFZ─Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich 81675, Germany
| | - Katja Steiger
- Comparative Experimental Pathology (CEP), School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan 85354, Germany
- EKFZ─Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich 81675, Germany
| | - Arne Skerra
- Chair of Biological Chemistry, School of Life Sciences, Technical University of Munich, Freising 85354, Germany
| |
Collapse
|
16
|
Diaz AV, Tekin I, Reis T. Drosophila as a Genetic Model System to Study Organismal Energy Metabolism. Biomolecules 2025; 15:652. [PMID: 40427545 PMCID: PMC12108566 DOI: 10.3390/biom15050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolism is the essential process by which an organism converts nutrients into energy to fuel growth, development, and repair. Metabolism at the level of a multicellular, multi-organ animal is inherently more complex than metabolism at the single-cell level. Indeed, each organ also must maintain its own homeostasis to function. At all three scales, homeostasis is a defining feature: as energy sources and energetic demands wax and wane, the system must be robust. While disruption of organismal energy homeostasis can be manifested in different ways in humans, obesity (defined as excess body fat) is an increasingly common outcome of metabolic imbalance. Here we will discuss the genetic basis of metabolic dysfunction that underlies obesity. We focus on what we are learning from Drosophila melanogaster as a model organism to explore and dissect genetic causes of metabolic dysfunction in the context of a whole organism.
Collapse
Affiliation(s)
| | | | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
17
|
Cassioli E, Lucherini Angeletti L, Rossi E, Selvi G, Riccardi E, Siviglia S, Buonanno R, Ricca V, Castellini G. Leptin Levels in Acute and Recovered Eating Disorders: An Arm-Based Network Meta-Analysis. EUROPEAN EATING DISORDERS REVIEW 2025; 33:525-537. [PMID: 39643920 PMCID: PMC11965547 DOI: 10.1002/erv.3163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE This study aimed to provide a BMI-adjusted meta-analytical calculation of blood leptin levels across different eating disorders (EDs) including anorexia nervosa (AN), bulimia nervosa (BN), binge eating disorder (BED), recovered EDs, and healthy controls (HCs). The goal was to understand BMI-independent leptin alterations and their potential as biomarkers. METHOD PubMed and ClinicalTrials.gov were searched for studies reporting serum leptin in AN, BN, BED, or recovered EDs. A multilevel network meta-analysis using a linear mixed-effects meta-regression model, adjusting for BMI, sex, and assay type, was performed on 146 studies (5048 patients, 3525 controls). RESULTS Significant differences in leptin levels were found across EDs. AN patients exhibited the lowest leptin levels, while BED patients had the highest. BN and recovered AN patients had leptin levels similar to AN, significantly lower than HCs. BMI, sex, and assay type were significant covariates. The model accounted for heterogeneity due to diagnostic criteria, assay types, and study-level differences. CONCLUSIONS Leptin levels in EDs are significantly altered beyond BMI effects, suggesting disease-specific factors. These findings support leptin's potential as a biomarker for ED staging and prognosis. Further research is needed to explore leptin's role in ED pathogenesis and trajectory, to identify subpopulations and improve clinical interventions.
Collapse
Affiliation(s)
- Emanuele Cassioli
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | | | - Eleonora Rossi
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Giulia Selvi
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Elena Riccardi
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Serena Siviglia
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Roberta Buonanno
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Valdo Ricca
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| | - Giovanni Castellini
- Psychiatry UnitDepartment of Health SciencesUniversity of FlorenceFlorenceItaly
| |
Collapse
|
18
|
Jahanbani A, Rezazadeh D, Sajadi E, Haj Hosseini M, Ketabchi D, EskandariRoozbahani N. Human adaptation response to obesity. Int J Obes (Lond) 2025:10.1038/s41366-025-01791-9. [PMID: 40287541 DOI: 10.1038/s41366-025-01791-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
This article examines the human body's adaptive responses to obesity from biological, behavioral, and evolutionary perspectives. It explores how ancient survival mechanisms, such as fat storage during scarcity, have persisted but become maladaptive in modern contexts of food abundance and sedentary lifestyles. Using the Thrifty Gene Hypothesis and General Adaptation Syndrome (GAS), the study investigates how chronic stress and genetic predispositions contribute to obesity. Chronic stress, as described in GAS, is linked to obesity through mechanisms like prolonged cortisol elevation, which promotes fat storage, particularly in the abdominal region, and disrupts hunger and satiety regulation. The article also explores the possibility that contemporary chronic stress may cause the body to buffer stressful conditions through fat accumulation. While the Thrifty Gene Hypothesis suggests that genetic traits evolved to optimize energy storage during scarcity, contributing to obesity in modern environments, it remains controversial. Critics argue that it oversimplifies obesity's causes, such as lifestyle and environmental factors. Although genetic variations influencing obesity susceptibility continue to evolve, the physiological mechanisms of fat storage and stress adaptation have remained largely unchanged since ancient times.
Collapse
Affiliation(s)
- Alireza Jahanbani
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Davood Rezazadeh
- Molecular Medicine Department, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elham Sajadi
- Department of Basic Science, Faculty of veterinary medicine, Shiraz University, Shiraz, Iran
| | - Mahdiyeh Haj Hosseini
- Department of Physical Education and Sport Sciences, National University of Skills (NS), Tehran, Iran
| | - Deniz Ketabchi
- School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Narges EskandariRoozbahani
- Clinical research development center, Imam Reza Hospital, Kermanshah University of Medical sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
van der Heide ME, Nørgaard JV, Madsen JG. Longitudinal pattern of growth performance and feeding behaviour in pigs fed varying dietary crude protein levels and categorised by final feed efficiency. Animal 2025; 19:101531. [PMID: 40414109 DOI: 10.1016/j.animal.2025.101531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/27/2025] Open
Abstract
Feed efficiency (FE) estimated on pen level does not take individual feed intake or feeding behaviour into account, and thus, varying response among pen mates is impossible to quantify. This study aimed to explore the within-pen variation in FE of pigs fed one of three levels of CP. The study included 60 gilts (30-115 kg) divided into two repetitions (10 pigs/pen with an automatic feeder). Pigs were fed a low (LCP; 120 g SID CP/kg), standard (SCP; 124 g SID CP/kg) or high (HCP; 128 g SID CP/kg) CP diet. Pigs within diet were categorised based on their FCR in the final four growing weeks as the 30% highest (HF), medium 40% (MF), and the 30% lowest (LF). Weekly BW, average daily gain (ADG), feed conversion ratio (FCR), and daily and per-meal feed intake and feeding behaviour were measured per individual. Data were analysed as polynomial mixed models. Growth performance parameters except FCR had significant dietary treatment (DT) × time (T) and feed conversion category × time interactions. During weeks (W) 10-12, BW tended (P ≤ 0.10) to be greater in HCP compared with LCP aligning with the numerically highest intake for HCP in the preceding period. Similarly, HF intermittently had greater BW and preceding higher intake than LF or MF (0.05 ≤ P ≤ 0.10). The ADG of LCP pigs showed the greatest variability over time and intermittently increased faster and slower (0.05 ≤ P ≤ 0.10) than SCP and HCP, respectively. From W10 onwards, the HF pigs had a stable ADG while the ADG of LF and MF increased steeply and consequently was greater (P ≤ 0.05) at W13-14. In line with ADG, FCR varied significantly over time and most strongly for LCP. Overall, LF pigs had the lowest, while HF pigs had the highest FCR (P < 0.001). Meal size (P < 0.001) and duration (P < 0.001) had a significant DT × T interaction, being greater for HCP than LCP and occasionally SCP. In line with differences in growth rate, meal frequency decreased faster (P < 0.05) for HCP than LCP or SCP, resulting in longer intervals and limited differences in daily feeding duration. While meal size and duration tended (P ≤ 0.10) to be greater for HF than MF or LF pigs, differences in daily feeding behaviour were negligible. It was concluded that, independent of CP-level, a distinction in FCR categories could be attributed to differential meal feeding behaviour of HF pigs leading to higher feed intake, whereas differences in ADG changed during the growing-finishing phase.
Collapse
Affiliation(s)
- M E van der Heide
- Department of Animal and Veterinary Sciences, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark
| | - J V Nørgaard
- Department of Animal and Veterinary Sciences, Aarhus University, Blichers Allé 20, 8830 Tjele, Denmark
| | - J G Madsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark.
| |
Collapse
|
20
|
Lambert GW, Patel M, Lambert EA. The Influence of the Sympathetic Nervous System on Cardiometabolic Health in Response to Weight Gain or Weight Loss. Metabolites 2025; 15:286. [PMID: 40422864 DOI: 10.3390/metabo15050286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Alterations in sympathetic nervous activity are evident in response to changes in body weight. Sympathetic nervous activity and sympathetic responses to weight change are regionalized, with alterations in end organ function dependent on the changes occurring in the brain regulatory pathways invoked and in the effector organs engaged. The obesity-induced activation of the sympathetic nervous system likely contributes to the initiation and worsening of cardiometabolic risk factors, including elevated blood pressure, cardiac dysfunction, dyslipidaemia, increased fasting blood glucose, insulin resistance, and non-alcoholic steatohepatitis. Unintended weight loss, as occurs in cachexia, is driven, at least in part, by the activation of sympathetic nervous-stimulated thermogenesis. The complexity of sympathetic nervous regulation renders the use of global measures of sympathetic activity problematic and the development of targeted therapies difficult, but these are not without promise or precedent. Knowledge of the central and peripheral pathways involved in sympathetic nervous regulation has opened up opportunities for pharmacological, surgical, and device-based approaches to mitigating the burden of disease development and progression. In this narrative review, we elaborate on sympathetic activity in response to changes in body weight, the brain pathways involved, and the cardiovascular and metabolic risks associated with perturbations in regional sympathetic activity.
Collapse
Affiliation(s)
- Gavin W Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Mariya Patel
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
| | - Elisabeth A Lambert
- School of Health Sciences and Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia
- Medical Technology Victoria (MedTechVic) Research Hub, Hawthorn, VIC 3122, Australia
| |
Collapse
|
21
|
Day KJ, Ballestro V, Mei L, Murgia C. Transcriptomic analysis of adipose tissue reveals adipogenesis is modulated by the degree of weight loss in patients with obesity. Nutr Metab Cardiovasc Dis 2025:104093. [PMID: 40345923 DOI: 10.1016/j.numecd.2025.104093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND AIMS Obesity is a complex condition with a diverse presentation and highly variable response to treatment. It is increasingly evident that obesity requires a plethora of interventions informed by biomarkers to optimise the chance of success. Adipose tissue is a metabolically active endocrine tissue and its metabolic response to intervention has the potential to provide key information to design targeted treatments to tackle obesity. To this aim, this study sought to identify, through transcriptomics analysis, common elements between adipose tissue of those undergoing bariatric surgery to those who were successful in losing weight through a lifestyle intervention. METHODS AND RESULTS Transcriptomic data from Gene Expression Omnibus were extracted from three bariatric surgery interventions and three lifestyle interventions. Paired linear mixed modelling using the limma package in R was used to determine differentially expressed genes before and after the interventions. Unpaired linear mixed modelling was used to determine differentially expressed genes between high and low responders to the intervention. Differentially modulated pathways were analysed with PathVisio and downloaded for graphic representation from the WikiPathways database. CONCLUSION We demonstrated that successful lifestyle interventions and bariatric surgery share the modulation of the Adipogenesis pathway (WP236). Leptin expression was positively correlated with weight loss, only up to a 10 % weight loss, suggesting a possible involvement of its dysregulation in explaining individual propensity to weight regain and potentially indicating who requires a targeted intervention. We also identify an interesting relationship between RBL2 expression and weight loss that requires further investigation.
Collapse
Affiliation(s)
- Kaitlin J Day
- School of Health and Biomedical Sciences, RMIT University, Australia.
| | - Vivian Ballestro
- School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Melbourne, Australia.
| | - Linghan Mei
- School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Melbourne, Australia.
| | - Chiara Murgia
- School of Agriculture, Food and Ecosystem Sciences, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
22
|
Solheim MH, Stroganov S, Chen W, Subagia PS, Bauder CA, Wnuk-Lipinski D, Del Río-Martín A, Sotelo-Hitschfeld T, Beddows CA, Klemm P, Dodd GT, Lundh S, Secher A, Wunderlich FT, Steuernagel L, Brüning JC. Hypothalamic PNOC/NPY neurons constitute mediators of leptin-controlled energy homeostasis. Cell 2025:S0092-8674(25)00403-9. [PMID: 40273910 DOI: 10.1016/j.cell.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 12/23/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
Leptin acts in the brain to suppress appetite, yet the responsible neurocircuitries underlying leptin's anorectic effect are incompletely defined. Prepronociceptin (PNOC)-expressing neurons mediate diet-induced hyperphagia and weight gain in mice. Here, we show that leptin regulates appetite and body weight via PNOC neurons, and that loss of leptin receptor (Lepr) expression in PNOC-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) causes hyperphagia and obesity. Restoring Lepr expression in PNOC neurons on a Lepr-null obese background substantially reduces body weight. Lepr inactivation in PNOC neurons increases neuropeptide Y (Npy) expression in a subset of hypothalamic PNOC neurons that do not express agouti-related peptide (Agrp). Selective chemogenetic activation of PNOC/NPY neurons promotes feeding to the same extent as activating all PNOCARC neurons, and overexpression of Npy in PNOCARC neurons promotes hyperphagia and obesity. Thus, we introduce PNOC/NPYARC neurons as an additional critical mediator of leptin action and as a promising target for obesity therapeutics.
Collapse
Affiliation(s)
- Marie H Solheim
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sima Stroganov
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Weiyi Chen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - P Sicilia Subagia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Corinna A Bauder
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Daria Wnuk-Lipinski
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Almudena Del Río-Martín
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Cait A Beddows
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Garron T Dodd
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, VIC, Australia
| | - Sofia Lundh
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Neurogenomics Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
23
|
Ohga H. Puberty regulation in chub mackerel Scomber japonicus, an important aquaculture fish species, via reproductive endocrine mechanism. Gen Comp Endocrinol 2025:114735. [PMID: 40254037 DOI: 10.1016/j.ygcen.2025.114735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
The vertebrate reproductive system is controlled by the brain-pituitary-gonadal reproductive endocrine axis (BPG axis). Gonadotropin-releasing hormone (GnRH) secreted from the hypothalamus regulates the secretion of two gonadotropic hormones (GTHs), follicle-stimulating hormone (FSH) and luteinizing hormone (LH), in the pituitary gland. Steroid hormones, such as androgens and estrogens, secreted in response to GTH stimulation, regulate the development of male and female gametes. Recently, various neuropeptides have been identified in mammals as factors that regulate the BPG axis from the upstream, demonstrating their importance. The author's main research theme is "Understanding the mechanism underlying puberty control in aquaculturally important species." They have been actively investigating the regulatory mechanism of the BPG axis in the puberty of chub mackerel Scomber japonicus, an important fishery species. With this species, researchers can systematically collect target organs, cells, and other organs from individuals at each developmental stage, from fertilized eggs to adult fish after spawning. A highly reproducible rearing experimental system has been established, enabling verification of the knowledge gained through feedback to reared individuals. Furthermore, it has recently become possible to quickly and efficiently produce gene knockout lines using genome editing. This article introduces the mechanism of puberty control in chub mackerel by fully leveraging this experimental platform. Focus will be directed to the functions of kisspeptin, which triggers puberty by regulating GnRH secretion in mammals, and leptin, which transmits nutritional information to the reproductive axis.
Collapse
Affiliation(s)
- Hirofumi Ohga
- Regional Fish Institute, Ltd., Fish Breeding Group, Aqua-plant Miyazu, 1001, Odashukuno, Miyazu-shi, Kyoto 626-0052, Japan
| |
Collapse
|
24
|
Jubayar AM, Khan S, Sadi J, Uddin N, Goni O, Khatun M, Siddique AE, Kabir E, Rimi RK, Beauty SA, Rossi KN, Abedin F, Sarder SJ, Islam MS, Sarker MK, Hossain S, Sumi D, Saud ZA, Barchowsky A, Wahed AS, Himeno S, Hossain K. Novel evidence of arsenic-related excess adiposity and its implication in the risk of cardiometabolic diseases. ENVIRONMENTAL RESEARCH 2025; 271:121059. [PMID: 39922254 PMCID: PMC11991875 DOI: 10.1016/j.envres.2025.121059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Arsenic exposure is associated with obesity- or excess adiposity-related disorders, including cardiometabolic diseases. Previously, many human studies attempted to establish the association of arsenic exposure with obesity, mainly through body mass index (BMI) but failed to provide any concrete evidence. Our study aimed to investigate the arsenic-related adiposity and its relationship with cardiometabolic diseases. Of the 524 participants, 126 and 398, respectively, were chosen from low- and high-arsenic exposure areas in Bangladesh. Obesity or body fat (adiposity) of the participants was measured by anthropometric measures [BMI, waist circumference (WC), and triceps skinfold thickness (TSFT)] and a serum biomarker, leptin. Sarcopenic characteristics were assessed by lean body mass (LBM) and serum creatinine levels. Insulin resistance, as measured by Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), hypertension, and hyperglycemia, were considered as the risk factors for cardiometabolic diseases. There were significant positive associations between drinking water, hair, and nail arsenic concentrations and the levels of WC and TSFT after adjusting for potential confounders. However, there were no significant associations with BMI. Increased arsenic exposure levels were associated with increased leptin levels [(Regression coefficient (β) = 1.00, 95% confidence interval (CI): 0.53, 1.46) for water, (β = 1.44, 95% CI: 0.42, 2.46) for hair, and (β = 1.47, 95% CI: 0.32, 2.61) for nail arsenic]. Notably, leptin levels had inverse associations with LBM (β = -7.87, 95% CI: -13.30, -2.45) and creatinine levels (β = -15.65, 95% CI: -21.50, -9.81). Furthermore, the elevated leptin levels associated with arsenic exposure were connected to higher HOMA-IR levels (β = 0.19, 95% CI: 0.14, 0.24), higher odds of hypertension [Odds ratio (OR) = 1.31, 95% CI: 1.12, 1.53], and hyperglycemia (OR = 1.30, 95% CI: 1.13, 1.47). Taken together, the results of this study demonstrated a unique association between arsenic exposure and adiposity, which could promote arsenic-induced cardiometabolic disorders by mirroring the distinctive characteristics of age-associated sarcopenic obesity.
Collapse
Affiliation(s)
- Ahsanul Mahbub Jubayar
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Shuchismita Khan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Junayed Sadi
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Nesar Uddin
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Osman Goni
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Moriom Khatun
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Abu Eabrahim Siddique
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, 52242, Iowa, USA
| | - Ehsanul Kabir
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Rajoana Karim Rimi
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sharmin Akter Beauty
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Kamrun Nahar Rossi
- Department of Pharmacy, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Faysal Abedin
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sharon Jahan Sarder
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Shofikul Islam
- Department of Applied Nutrition and Food Technology, Islamic University, Kushtia, 7003, Bangladesh
| | | | - Shakhawoat Hossain
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Daigo Sumi
- Laboratory of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan
| | - Zahangir Alam Saud
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Abdus S Wahed
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Seiichiro Himeno
- Laboratory of Molecular Toxicology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, 770-8514, Japan; Division of Health Chemistry, School of Pharmacy, Showa University, Tokyo, 142-8555, Japan
| | - Khaled Hossain
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
25
|
Lupu A, Mihai CM, Dragan F, Tarnita I, Alecsa M, Chisnoiu T, Morariu ID, Cuciureanu M, Nedelcu AH, Salaru DL, Anton E, Danielescu C, Fotea S, Stoleriu G, Beser OF, Lupu VV. Antioxidant Supplementation in Childhood Obesity: A Path to Improved Metabolic Health? Antioxidants (Basel) 2025; 14:466. [PMID: 40298814 PMCID: PMC12024302 DOI: 10.3390/antiox14040466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Childhood obesity is linked to heightened oxidative stress, a key driver of endothelial dysfunction, inflammation, and metabolic complications. Antioxidants, including Vitamins C and E, are vital in neutralizing free radicals and mitigating oxidative damage. This non-systematic review examines the potential advantages of antioxidant supplementation in pediatric obesity, focusing on its effects on vascular health, insulin sensitivity, and inflammatory processes. Emerging data suggest that antioxidants may improve endothelial function, reduce blood pressure, and enhance metabolic homeostasis in obese children. However, the long-term efficacy and safety of antioxidant supplementation remain uncertain, necessitating further rigorous randomized controlled trials. A deeper understanding of antioxidants' role in pediatric obesity could unlock novel therapeutic approaches for managing obesity-related complications and improving children's overall health outcomes.
Collapse
Affiliation(s)
- Ancuta Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Cristina Maria Mihai
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Felicia Dragan
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Irina Tarnita
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Mirabela Alecsa
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| | - Tatiana Chisnoiu
- Pediatrics, Faculty of Medicine, “Ovidius” University, 900470 Constanta, Romania; (C.M.M.); (T.C.)
| | - Ionela Daniela Morariu
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Magdalena Cuciureanu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Alin Horatiu Nedelcu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Delia Lidia Salaru
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Emil Anton
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Ciprian Danielescu
- Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.C.); (A.H.N.); (D.L.S.); (E.A.); (C.D.)
| | - Silvia Fotea
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (S.F.); (G.S.)
| | - Gabriela Stoleriu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800008 Galati, Romania; (S.F.); (G.S.)
| | - Omer Faruk Beser
- Department of Pediatric Gastroenterology, Hepatology & Nutrition, Cerrahpasa Medical Faculty, Istanbul University Cerrahpasa, 34776 Istanbul, Turkey;
| | - Vasile Valeriu Lupu
- Pediatrics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (A.L.); (M.A.); (V.V.L.)
| |
Collapse
|
26
|
Moura-Assis A, Velloso LA. Leptin 30 years - A chat with Jeffrey M. Friedman. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2025; 68:e240413. [PMID: 40215468 PMCID: PMC11967189 DOI: 10.20945/2359-4292-2024-0413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 04/15/2025]
Affiliation(s)
| | - Licio A. Velloso
- Centro de Pesquisa em Obesidade e Comorbidades, Universidade Estadual de
Campinas, Campinas, SP, Brasil
- Instituto Nacional de Ciência e Tecnologia em
Neuroimunomodulação, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
27
|
Catana OM, Nemes AF, Cioboata R, Toma CL, Mitroi DM, Calarasu C, Streba CT. Leptin and Insulin in COPD: Unveiling the Metabolic-Inflammatory Axis-A Narrative Review. J Clin Med 2025; 14:2611. [PMID: 40283443 PMCID: PMC12027990 DOI: 10.3390/jcm14082611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive and debilitating condition characterized by airflow limitations and systemic inflammation. The interaction between the metabolic and inflammatory pathways plays a key role in disease progression, with leptin and insulin emerging as pivotal metabolic regulators. Leptin, an adipokine that regulates energy homeostasis, and insulin, the primary regulator of glucose metabolism, are both altered in COPD patients. This narrative review provides an in-depth examination of the roles of leptin and insulin in COPD pathogenesis, focusing on the molecular mechanisms through which these metabolic regulators interact with inflammatory pathways and how their dysregulation contributes to a spectrum of extrapulmonary manifestations. These disturbances not only exacerbate COPD symptoms but also increase the risk of comorbidities such as metabolic syndrome, diabetes, cardiovascular disease, or muscle wasting. By exploring the underlying mechanisms of leptin and insulin dysregulation in COPD, this review underscores the significance of the metabolic-inflammatory axis, suggesting that restoring metabolic balance through leptin and insulin modulation could offer novel therapeutic strategies for improving clinical outcomes.
Collapse
Affiliation(s)
- Oana Maria Catana
- Doctoral School, University of Medicine and Pharmacy, 200349 Craiova, Romania; (O.M.C.); (D.M.M.)
| | | | - Ramona Cioboata
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania; (C.C.); (C.T.S.)
| | - Claudia Lucia Toma
- Pneumology Department, University of Medicine Carol Davila, 020021 Bucharest, Romania
| | - Denisa Maria Mitroi
- Doctoral School, University of Medicine and Pharmacy, 200349 Craiova, Romania; (O.M.C.); (D.M.M.)
| | - Cristina Calarasu
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania; (C.C.); (C.T.S.)
| | - Costin Teodor Streba
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania; (C.C.); (C.T.S.)
| |
Collapse
|
28
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
29
|
Blüher M. An overview of obesity-related complications: The epidemiological evidence linking body weight and other markers of obesity to adverse health outcomes. Diabetes Obes Metab 2025; 27 Suppl 2:3-19. [PMID: 40069923 PMCID: PMC12000860 DOI: 10.1111/dom.16263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/23/2025] [Accepted: 02/02/2025] [Indexed: 04/17/2025]
Abstract
Obesity is a highly prevalent chronic multisystem disease associated with shortened life expectancy due to a number of adverse health outcomes. Epidemiological data link body weight and parameters of central fat distribution to an increasing risk for type 2 diabetes, hypertension, fatty liver diseases, cardiovascular diseases including myocardial infarction, heart failure, atrial fibrillation, stroke, obstructive sleep apnoea, osteoarthritis, mental disorders and some types of cancer. However, the individual risk to develop cardiometabolic and other obesity-related diseases cannot entirely be explained by increased fat mass. Rather than excess fat accumulation, dysfunction of adipose tissue may represent the mechanistic link between obesity and adverse health outcomes. There are people living with obesity who seem to be protected against the premature development of cardiometabolic diseases. On the other hand, people with normal weight may develop typical obesity diseases upon dysfunction of adipose tissue and predominantly visceral fat distribution. The mechanisms linking impaired function of adipose tissue in people with obesity include adipocyte hypertrophy, altered cellular composition, limited expandability of safe subcutaneous fat stores, ectopic fat deposition in visceral depots, the liver and other organs, hypoxia, a variety of stresses, inflammatory processes, and the release of pro-inflammatory, diabetogenic and atherogenic signals. Genetic and environmental factors might contribute either alone or via interaction with intrinsic biological factors to variation in adipose tissue function. There are still many open questions regarding the mechanisms of how increased body weight causes obesity-related disorders and whether these pathologies could be reversed. Evidence-based weight loss interventions using behaviour change, pharmacological or surgical approaches have clarified the beneficial effects of realistic and sustained weight loss on obesity-related complications as hard outcomes. This review focusses on recent advances in understanding epidemiological trends and mechanisms of obesity-related diseases. PLAIN LANGUAGE SUMMARY: Obesity is a chronic complex and progressive disease characterized by excessive fat deposition that may impair health and quality of life. Worldwide, the number of adults living with obesity has more than doubled since 1990. Obesity may lead to reduced life expectancy, because it increases the risk for type 2 diabetes, cardiovascular diseases (e.g., myocardial infarction, high blood pressure, stroke), fatty liver diseases, musculoskeletal diseases, chronic respiratory diseases, depression and certain types of cancer. However, not every person with obesity develops these diseases. For better prevention and treatment, it is important to understand the mechanisms linking high fat mass to obesity related diseases. It has become clear that fat mass alone cannot explain the higher risk of obesity complications. People with obesity can have either high or low risk of developing complications. Compared to people with a low risk for obesity complications those with a high risk to develop obesity related diseases are characterized by higher central fat deposition in the abdominal region, on average bigger fat cells, higher number of immune cells in adipose tissue and altered signals released from adipose tissue that may directly affect the brain, liver, vasculature and other organs. Both inherited and environment factors may cause these abnormalities of adipose tissue function. However, weight loss through behaviour changes (e.g., lower calorie intake, higher physical activity), medications or obesity surgery can improve health, quality of life and reduce the risk for obesity related diseases.
Collapse
Affiliation(s)
- Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum MünchenUniversity of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III—Endocrinology, Nephrology, RheumatologyUniversity of Leipzig Medical CenterLeipzigGermany
| |
Collapse
|
30
|
Lin Z, Xuan Y, Zhang Y, Zhou Q, Qiu W. Hypothalamus and brainstem circuits in the regulation of glucose homeostasis. Am J Physiol Endocrinol Metab 2025; 328:E588-E598. [PMID: 40047236 DOI: 10.1152/ajpendo.00474.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/03/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
The central nervous system (CNS) senses and integrates blood glucose status, regulating its levels through communication with peripheral organs. Since traditional wisdom holds that the hypothalamus primarily controls glucose homeostasis, the brainstem, although less studied, has been emerging as a key player in blood glucose metabolism. Although the brainstem is reciprocally wired with the hypothalamus, their interactions are crucial for glucose control. Here, we focus on classic discoveries and recent advancements of hypothalamic and brainstem nodes that regulate glucose homeostasis. Based on the current progress and development for central regulation of blood sugar, we propose that the circuitry and cellular mechanisms for how hypothalamus and brainstem coordinate in blood sugar regulation are crucial; hence, a deeper understanding of both nuclei could shed light on a future cure for diabetes.
Collapse
Affiliation(s)
- Zitian Lin
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yunxin Xuan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Yingshi Zhang
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Qirui Zhou
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| | - Weiwei Qiu
- Department of Endocrinology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, People's Republic of China
| |
Collapse
|
31
|
Xu DL, Hu XK. Effect of housing density on cellular and humoral immunity, hematology in striped hamsters. J Comp Physiol B 2025; 195:235-245. [PMID: 39992410 DOI: 10.1007/s00360-025-01605-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Population density is one of the most important factors influencing immune function. Social stress induced by higher density may account for the immunosuppression according to the endocrine hypothesis. To test this hypothesis, male striped hamsters (Cricetulus barabensis) were classified into the One/Cage (n = 9), Two/Cage (n = 6), and Three/Cage (n = 9) groups, and the treatment lasted for 45 days. The titers of immunoglobin (Ig)G15 and IgM 10 were lower in the Two/Cage group compared to the One/Cage group, indicating that higher housing density suppressed humoral immunity. However, the masses of thymus and spleen, phytohaemagglutinin (PHA) responses at 6 h, 12 h, 24 h, 48 h, and 72 h after PHA injection, the titers of IgG 5, IgG10, IgM5, and IgM15 were all not affected by housing density. Blood glucose level was higher in the One/Cage group than the other two groups, leptin titers did not differ among the three groups, whereas corticosterone concentration was higher in the Two/Cage and Three/Cage groups than in the One/Cage group. Moreover, negative correlation was observed between corticosterone concentration and the titers of IgG5, IgG10, IgG15, and IgM10. These results suggested that humoral immunity was reduced by higher stress levels induced by higher housing density, which supported the endocrine hypothesis. White blood cell (WBC) count was higher in the Two/Cage group than in the One/Cage group, and intermediate granulocytes (MID) were higher in the Two/Cage group than in the One/Cage and Three/Cage groups, indicating the fight and injury might have arisen in the higher housing density.
Collapse
Affiliation(s)
- De-Li Xu
- School of Life Sciences, Qufu Normal University, 57 Jingxuan West Road, Qufu, Shandong Province, 273165, People's Republic of China.
| | - Xiao-Kai Hu
- School of Life Sciences, Qufu Normal University, 57 Jingxuan West Road, Qufu, Shandong Province, 273165, People's Republic of China
| |
Collapse
|
32
|
Bilgiç A, Cura M, Kılınç İ, Akça ÖF. Low Levels of Serum Ghrelin and Nesfatin-1 Are Associated With Anxiety Disorders in Children. Soa Chongsonyon Chongsin Uihak 2025; 36:69-77. [PMID: 40203141 PMCID: PMC11969048 DOI: 10.5765/jkacap.250001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Objectives Because appetite-regulating hormones are implicated in neuronal survival, growth, and differentiation, they have been suggested to play a role in anxiety disorders. To date, few studies have focused on the association between these hormones and anxiety disorders in children. This study investigated the potential differences in leptin, ghrelin, and nesfatin-1 serum levels in drug-naïve children with anxiety disorders, including social anxiety disorder, separation anxiety disorder, and generalized anxiety disorder, and in healthy controls. Methods This study included 45 children (14 boys and 31 girls) with anxiety disorders and 35 healthy controls (13 boys and 22 girls) aged 8-18 years. The severity of anxiety disorders and additional symptoms were evaluated using the Revised Child Anxiety and Depression Scales-Child Version. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate leptin, ghrelin, and nesfatin-1 serum levels. Results Leptin levels were significantly higher in children with anxiety disorders than in the control group, and ghrelin and nesfatin-1 levels were significantly lower in children with anxiety disorders than in the control group for girls and for the entire sample. However, only low nesfatin-1 levels were significantly associated with anxiety disorders in boys. In the entire sample, potential confounders such as age, sex, body mass index, and the severity of depressive symptoms were controlled for, and the results were the same for ghrelin and nesfatin-1 levels. However, the difference in leptin levels between groups was not significant. Conclusion These findings suggest that dysregulation of ghrelin and nesfatin-1 concentrations may be related to the etiopathogenesis of childhood anxiety disorders.
Collapse
Affiliation(s)
- Ayhan Bilgiç
- Department of Child and Adolescent Psychiatry, Faculty of
Medicine, Izmir University of Economics, Izmir,
Türkiye
| | - Merve Cura
- Department of Child and Adolescent Psychiatry, Etlik City
Hospital, Ankara, Türkiye
| | - İbrahim Kılınç
- Department of Biochemistry, Necmettin Erbakan University
Faculty of Medicine, Konya, Türkiye
| | - Ömer Faruk Akça
- Department of Child and Adolescent Psychiatry, Necmettin
Erbakan University Faculty of Medicine, Konya, Türkiye
| |
Collapse
|
33
|
Liu C, Yu J, Du Y, Xie Y, Song X, Liu C, Yan Y, Wang Y, Qin J. Synergistic Activation of LEPR and ADRB2 Induced by Leptin Enhances Reactive Oxygen Specie Generation in Triple-Negative Breast Cancer Cells. Cancer Res Treat 2025; 57:457-477. [PMID: 39164083 PMCID: PMC12016824 DOI: 10.4143/crt.2024.368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/19/2024] [Indexed: 08/22/2024] Open
Abstract
PURPOSE Leptin interacts not only with leptin receptor (LEPR) but also engages with other receptors. While the pro-oncogenic effects of the adrenergic receptor β2 (ADRB2) are well-established, the role of leptin in activating ADRB2 in triple-negative breast cancer (TNBC) remains unclear. MATERIALS AND METHODS The pro-carcinogenic effects of LEPR were investigated using murine TNBC cell lines, 4T1 and EMT6, and a tumor-bearing mouse model. Expression levels of LEPR, NADPH oxidase 4 (NOX4), and ADRB2 in TNBC cells and tumor tissues were analyzed via western blot and quantitative real-time polymerase chain reaction. Changes in reactive oxygen species (ROS) levels were assessed using flow cytometry and MitoSox staining, while immunofluorescence double-staining confirmed the co-localization of LEPR and ADRB2. RESULTS LEPR activation promoted NOX4-derived ROS and mitochondrial ROS production, facilitating TNBC cell proliferation and migration, effects which were mitigated by the LEPR inhibitor Allo-aca. Co-expression of LEPR and ADRB2 was observed on cell membranes, and bioinformatics data revealed a positive correlation between the two receptors. Leptin activated both LEPR and ADRB2, enhancing intracellular ROS generation and promoting tumor progression, which was effectively countered by a specific ADRB2 inhibitor ICI118551. In vivo, leptin injection accelerated tumor growth and lung metastases without affecting appetite, while treatments with Allo-aca or ICI118551 mitigated these effects. CONCLUSION This study demonstrates that leptin stimulates the growth and metastasis of TNBC through the activation of both LEPR and ADRB2, resulting in increased ROS production. These findings highlight LEPR and ADRB2 as potential biomarkers and therapeutic targets in TNBC.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Jing Yu
- School of Medicine, Nankai University, Tianjin, China
| | - Yongjun Du
- School of Medicine, Nankai University, Tianjin, China
| | - Yu Xie
- School of Medicine, Nankai University, Tianjin, China
| | - Xiaofei Song
- School of Medicine, Nankai University, Tianjin, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Yan Yan
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Yue Wang
- School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Junfang Qin
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
34
|
Falkenhain K, De Baca TC, Stinson EJ, Ravussin E, Piaggi P, Krakoff J, Redman LM. The effect of a leptin phenotype on weight change and energy expenditure responses to acute and prolonged energetic stressors. Am J Physiol Endocrinol Metab 2025; 328:E579-E587. [PMID: 40094227 PMCID: PMC12067472 DOI: 10.1152/ajpendo.00067.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Leptin is a hormone produced by adipocytes that plays a crucial role in regulating energy homeostasis and body mass. Despite its close correlation with body fat, up to ∼40% of variation in plasma leptin concentration remains unexplained, allowing for the classification of a distinct "leptin phenotype." This leptin phenotype-characterized by either relatively high or relatively low leptin concentration relative to an individual's level of body fat-presents an intriguing opportunity to test whether relatively higher (compared with lower) leptin concentrations differentially affect energy expenditure, metabolic adaptation, and susceptibility to weight change in response to energy balance perturbations. To test this hypothesis, we characterized the energy expenditure and weight change response between the two leptin phenotypes (relatively high vs. low) using three distinct experimental contexts: a cross-sectional analysis (n = 104), acute (24-h) perturbations with fasting and overfeeding (n = 77), and chronic perturbations with 24-mo caloric restriction (n = 144) or 8-wk overfeeding (n = 28). Leptin phenotype did not explain variations in energy expenditure responses either in cross-sectional analyses or in response to acute or prolonged energetic stressors. Moreover, leptin phenotype was not a determinant of weight change in response to energy restriction or surplus, or subsequent weight recovery. These results suggest that classifying individuals based on a leptin phenotype does not allow to detect differential susceptibility to energy expenditure adaptations or weight change.NEW & NOTEWORTHY Leptin is linked to body fat, but unexplained variation remains. This study challenges the idea that distinct leptin phenotypes-characterized by relatively high or low leptin concentration for a given level of body fat-affects energy expenditure or weight change in response to acute or prolonged energy stressors. We found no association between leptin phenotypes and energy expenditure or weight change either cross-sectionally or in response to acute or prolonged over- or underfeeding.
Collapse
Affiliation(s)
- Kaja Falkenhain
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Tomás Cabeza De Baca
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Emma J Stinson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Paolo Piaggi
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Phoenix, Arizona, USA
| | - Leanne M Redman
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
35
|
Li Z, Ma Q, Zhang J, Yin R, You J, Hao Q, Wu X, Kang J, Wang L, Deng Y, Li Y, Shen C, Wu B, Feng J, Tu Y, Xiao X, Yu J, Cheng W. Large-Scale Plasma Proteomics to Profile Pathways and Prognosis of Chronic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410160. [PMID: 40048323 PMCID: PMC12021123 DOI: 10.1002/advs.202410160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/10/2025] [Indexed: 04/26/2025]
Abstract
While increasing peripheral mechanisms related to chronic pain, the plasma proteomics profile associated with it and its prognosis remains elusive. This study utilizes 2923 plasma proteins and chronic pain of 51 644 participants from UK Biobank and finds 474 proteins linked to chronic pain in six sites: head, neck or shoulder, back, stomach or abdominal, hip, and knee, with 11 proteins sharing across pain sites. The identified proteins are largely enriched in immune and metabolic pathways and highly expressed in tissues like lungs and small intestines. Phenome-wide analysis highlights the significance of pain-related proteome on diverse facets of human health, and in-depth Mendelian randomization validates 10 proteins (CD302, RARRES2, TNFRSF1B, BTN2A1, TNFRSF9, COL18A1, TNF, CD74, TNFRSF4, and BTN2A1) as markers of chronic pain. Furthermore, protein sets capable of classifying pain patients and healthy participants, particularly performing best in hip pain (area under curve, AUC = 0.725), are identified. Interestingly, the prediction of pain spreading over ten years achieves an AUC of 0.715, with leptin identified as a crucial predictor. This study delineates proteins associated with various pain conditions and identifies proteins capable of classifying pain and predicting pain spreading, offering benefits for both research and clinical practice.
Collapse
Affiliation(s)
- Ze‐Yu Li
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Qing Ma
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
- Key Laboratory of Brain Functional Genomics (MOE&STCSM), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive ScienceEast China Normal UniversityShanghai200062China
| | - Jie Zhang
- Department of NeurosurgeryHuashan Hospital, Shanghai Medical CollegeFudan UniversityShanghai200040China
- National Center for Neurological DisordersShanghai200040China
| | - Rui‐Ying Yin
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Jia You
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Qi‐Zheng Hao
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Xin‐Rui Wu
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
| | - Ju‐Jiao Kang
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Lin‐Bo Wang
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Yue‐Ting Deng
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
| | - Yu‐Zhu Li
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Chun Shen
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
| | - Bang‐Sheng Wu
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
| | - Jian‐Feng Feng
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
- Department of Computer ScienceUniversity of WarwickCoventryCV4 7ALUK
- Fudan ISTBI–ZJNU Algorithm Centre for Brain‐inspired IntelligenceZhejiang Normal UniversityZhejiang321004China
| | - Yi‐Heng Tu
- CAS Key Laboratory of Mental Health, Institute of PsychologyChinese Academy of SciencesBeijing100101China
| | - Xiao Xiao
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
| | - Jin‐Tai Yu
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
| | - Wei Cheng
- Institute of Science and Technology for Brain‐Inspired IntelligenceDepartment of NeurologyHuashan HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200433China
- Key Laboratory of Computational Neuroscience and Brain‐Inspired Intelligence (Fudan University)Ministry of EducationShanghai200433China
- Fudan ISTBI–ZJNU Algorithm Centre for Brain‐inspired IntelligenceZhejiang Normal UniversityZhejiang321004China
| |
Collapse
|
36
|
Beeghly GF, Pincus MI, Varshney RR, Giri DD, Falcone DJ, Rudolph MC, Antonyak MA, Iyengar NM, Fischbach C. Hypertrophic adipocytes increase extracellular vesicle-mediated lipid release and reprogram breast cancer cell metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645549. [PMID: 40236195 PMCID: PMC11996363 DOI: 10.1101/2025.03.28.645549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Obesity worsens cancer-specific survival and all-cause mortality for women diagnosed with breast cancer. Rich in adipose tissue, the breast exhibits increased adipocyte size in obesity, which correlates with poor prognosis. However, adipocyte size is highly heterogeneous as adipose tissue expands through both hyperplasia and hypertrophy; and adipocyte size can increase independently of weight gain. Despite these observations, the impact of adipocyte size on breast cancer cell behavior remains unclear due to insufficient approaches to isolate adipocytes based on size and maintain them in culture for mechanistic studies. Here, we develop strategies to culture size-sorted adipocytes from two mouse models of obesity and test their functional impact on tumor cell malignancy. We find that large adipocytes are transcriptionally distinct from small adipocytes and are enriched for gene sets related to adipose tissue dysfunction, including altered lipid processing. In coculture studies, large adipocytes promote lipid accumulation in breast cancer cells, and enhance their migration, proliferation, and aerobic metabolism in a manner dependent on fatty acid oxidation. These changes coincide with increased release of extracellular vesicles by large versus small adipocytes, which transfer lipid to recipient tumor cells. Moving forward, our findings suggest that adipocyte size could serve as a prognostic biomarker for women with breast cancer and help identify new therapeutic targets to advance clinical outcomes for these patients.
Collapse
|
37
|
Mancini M, Hikima T, Witkovsky P, Patel JC, Stone DW, Affinati AH, Rice ME. Leptin activates dopamine and GABA neurons in the substantia nigra via a local pars compacta-pars reticulata circuit. J Neurosci 2025; 45:e1539242025. [PMID: 40127936 PMCID: PMC12096038 DOI: 10.1523/jneurosci.1539-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/28/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025] Open
Abstract
Adipose-derived leptin contributes to energy homeostasis by balancing food intake and motor output, but how leptin acts in brain motor centers remains poorly understood. We investigated the influence of leptin on neuronal activity in two basal ganglia nuclei involved in motor control: the substantia nigra pars compacta (SNc) and pars reticulata (SNr). Using a mouse reporter line to identify cells expressing leptin receptors (LepRs), we found that in both sexes, a majority of SNc dopamine neurons express a high level of LepR. Whole-cell recording in ex vivo midbrain slices from male wild-type mice showed that leptin activates SNc dopamine neurons directly and increases somatodendritic dopamine release. Although LepR expression in SNr GABA output neurons was low, leptin also activated these cells. Additional experiments showed that the influence of leptin on SNr neurons is indirect and involves D1 dopamine receptors and TRPC3 channels. Administration of leptin to male mice increased locomotor activity, consistent with activation of dopamine neurons in the SNc coupled to previously reported amplification of axonal dopamine release by leptin in striatal slices. These findings indicate that in addition to managing energy homeostasis through its actions as a satiety hormone, leptin also promotes axonal and somatodendritic dopamine release that can influence motor output.Significance statement Dopamine neurons regulate motivated behaviors, but how they are influenced by metabolic hormones, like leptin, is incompletely understood. We show here that leptin increases the activity of substantia nigra (SN) pars compacta dopamine neurons directly, and that this enhances somatodendritic dopamine release. Leptin also increases the activity of GABAergic neurons in the SN pars reticulata, but does so indirectly via D1 dopamine receptors activated by locally released dopamine. Consistent with increased nigral dopamine neuron activity and previous evidence showing that leptin amplifies striatal dopamine release, systemic leptin increases locomotor behavior. This increase in motor activity complements the well-established inhibitory effect of leptin on food intake and adds an additional dimension to the regulation of energy balance by this hormone.
Collapse
Affiliation(s)
- Maria Mancini
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, USA
- Neuroscience Institute, NYU School of Medicine, New York, NY, USA
| | - Takuya Hikima
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Paul Witkovsky
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Jyoti C Patel
- Neuroscience Institute, NYU School of Medicine, New York, NY, USA
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Dominic W Stone
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Alison H Affinati
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Margaret E Rice
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, USA
- Neuroscience Institute, NYU School of Medicine, New York, NY, USA
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
38
|
Lord RA, Inglis MA, Juengel JL, Anderson GM. A Leptin Receptor Mutation Which Impairs Fertility in Ewes Causes Delayed Puberty in Male and Female Mice. Endocrinology 2025; 166:bqaf058. [PMID: 40130278 PMCID: PMC11979093 DOI: 10.1210/endocr/bqaf058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 03/26/2025]
Abstract
Reproductive function is tightly linked to nutritional status due to its high energetic demands. Leptin, a key adipose tissue-derived hormone signalling energy reserves to the brain, integrates metabolic status with the hypothalamic-pituitary-gonadal axis to ensure reproductive function is maintained or suppressed appropriately. Mutations in leptin or its receptor (LepR) are known to cause infertility and obesity in mice. In Davisdale ewes, 2 naturally occurring LepR mutations (R62C and P1019S) were associated with delayed puberty and subfertility, but their effects in males or in other species remain to be determined. This study examined the impact of analogous LepR mutations (A63C and P1018S) in mice using CRISPR-Cas9 gene editing. Puberty onset, adult fertility, and metabolic phenotypes were assessed in wild-type, heterozygous, and homozygous mutant mice. The A63C mutation, located in the extracellular domain of the receptor, resulted in increased body weight and adiposity in females, along with delays in puberty onset in both sexes. Despite these delays, adult reproductive function was maintained. Immunohistochemical analysis revealed no detectable reductions in leptin-induced pSTAT3, pERK1/2, or pmTOR signalling in the hypothalamic arcuate nucleus in either mutant line, indicating these pathways remain largely intact. These findings demonstrate the conserved importance of this region of the leptin receptor for puberty onset and adiposity across species, but also the resilience of leptin signalling in preserving reproductive function despite genetic variation.
Collapse
Affiliation(s)
- Rebecca A Lord
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Megan A Inglis
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| | - Jennifer L Juengel
- Agricultural Systems and Reproduction, AgResearch Ltd, Invermay Agricultural Centre, Mosgiel 9092, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology, and Department of Anatomy, University of Otago School of Biomedical Sciences, Dunedin 9016, New Zealand
| |
Collapse
|
39
|
Villagrán-Silva F, Loren P, Sandoval C, Lanas F, Salazar LA. Circulating microRNAs as Potential Biomarkers of Overweight and Obesity in Adults: A Narrative Review. Genes (Basel) 2025; 16:349. [PMID: 40149500 PMCID: PMC11942292 DOI: 10.3390/genes16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
In an obesogenic environment, such as the one we have been experiencing in recent decades, epigenetics provides answers to the relationship between hereditary and environmentally acquired patterns that have significantly contributed to the global rise in obesity prevalence. MicroRNA (miRNA) constitutes a diminutive non-coding small RNA molecule, 20 to 24 nucleotides in length, that functions as a regulator of gene regulation at the post-translational level. Circulating miRNAs (c-miRNAs) have been detected in multiple body fluids, including blood, plasma, serum, saliva, milk from breastfeeding mothers, and urine. These molecules hold significant therapeutic value and serve as extracellular biomarkers in metabolic diseases. They aid in the diagnosis and tracking of therapy responses, as well as dietary and physical habit modifications. Researchers have studied c-miRNAs as potential biomarkers for diagnosing and characterizing systemic diseases in people of all ages and backgrounds since then. These conditions encompass dyslipidemia, type 2 diabetes mellitus (T2DM), cardiovascular risk, metabolic syndrome, cardiovascular diseases, and obesity. This review therefore analyzes the usefulness of c-miRNAs as therapeutic markers over the past decades. It also provides an update on c-miRNAs associated with general obesity and overweight, as well as with the most prevalent pathologies in the adult population. It also examines the effect of different nutritional approaches and physical activity regarding the activity of miRNAs in circulation in adults with overweight or general obesity. All of this is done with the aim of evaluating their potential use as biomarkers in various research contexts related to overweight and obesity in adults.
Collapse
Affiliation(s)
- Francisca Villagrán-Silva
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile;
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Fernando Lanas
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| |
Collapse
|
40
|
Grajales-Reyes JG. Advances in energy balance & metabolism circuitry. ADVANCES IN GENETICS 2025; 113:1-28. [PMID: 40409794 DOI: 10.1016/bs.adgen.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Advancements in informatics, genetics, and neuroscience have greatly expanded our understanding of how the central nervous system (CNS) regulates energy balance and metabolism. This chapter explores the key neural circuits within the hypothalamus and brainstem that integrate behavioral and physiological processes to maintain metabolic homeostasis. It also examines the dynamic interplay between the CNS and peripheral organs, mediated through hormonal and neuronal signals, which fine-tune appetite, energy expenditure, and body weight. Furthermore, we highlight groundbreaking research that unveils molecular and cellular pathways governing energy regulation, representing a new frontier in addressing obesity and metabolic disorders. Innovative approaches, such as neurogenetic and neuromodulation techniques, are explored as promising strategies for improving weight management and metabolic health. By providing a comprehensive perspective on the mechanisms underlying energy balance, this chapter underscores the transformative potential of emerging therapeutic innovations.
Collapse
Affiliation(s)
- Jose G Grajales-Reyes
- Department of Anesthesiology, Yale University, New Haven, CT, United States; Laboratory of Neurovascular Control of Homeostasis, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
41
|
Friedman JM. On the causes of obesity and its treatment: The end of the beginning. Cell Metab 2025; 37:570-577. [PMID: 40043689 DOI: 10.1016/j.cmet.2025.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 05/13/2025]
Abstract
Over the last 30 years, our understanding of the causes of obesity has been transformed, and new, highly effective medicines for reducing weight have been developed. This remarkable progress marks an end and a beginning. By establishing that obesity is a biologic disorder amenable to scientific inquiry and rational drug development, simplistic notions about its causes and treatment should be laid to rest. The future holds the promise that additional therapeutic approaches for inducing or maintaining weight loss will be developed, and that these treatments will be tailored to different subgroups to potentially address the pathogenic mechanisms.
Collapse
Affiliation(s)
- Jeffrey M Friedman
- Rockefeller University and Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
42
|
Ouyang P, Qi J, Tong B, Li Y, Cao J, Wang L, Niu T, Qi X. Butyrate Ameliorates Graves' Orbitopathy Through Regulating Orbital Fibroblast Phenotypes and Gut Microbiota. Invest Ophthalmol Vis Sci 2025; 66:5. [PMID: 40035727 PMCID: PMC11892527 DOI: 10.1167/iovs.66.3.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Purpose Graves' orbitopathy (GO), the common extrathyroidal complication of Graves' disease (GD), is characterized by orbital fibroblast stimulation, adipogenesis, and hyaluronan production. Recently, gut microbiota and its metabolites have garnered attention for their possible involvement in GO. Methods This study utilized an animal model of GO and examined the effects of butyrate treatment on orbital fibroblast cells and gut microbiota. Ex vivo experiments were performed using orbital fibroblasts derived from healthy patients' and patients' with GO orbital tissue to evaluate vitality, activation, and adipogenesis in response to butyrate treatment. Gut microbiota diversity was also analyzed in butyrate-treated and untreated GO mice. Results In human orbital fibroblasts, butyrate treatment dramatically decreased the vitality of GO-derived fibroblasts without harming normal fibroblasts. Butyrate prevented activation and fibrotic processes induced by transforming growth factor beta 1 (TGF-β1) in GO and normal fibroblasts. Additionally, butyrate reduced lipid droplet formation and downregulated lipogenic markers in GO and normal orbital fibroblasts, inhibiting adipogenesis. In the GO mouse model, butyrate therapy improved orbital histological abnormalities and normalized serum thyroid hormone and antibody levels. The intestinal microbiome of butyrate-treated GO mice also changed significantly, with a reduction in certain bacteria (Bifidobacterium, GCA-900066575, and Parabacteroides) and an increase in others (Bacteroides and Rikenellaceae_RC9). Conclusions Butyrate ameliorates several of the symptoms of GO, lowering GO orbital fibroblast viability, adipogenesis, and TGF-β1-induced fibrosis without damaging normal fibroblasts. Butyrate normalizes thyroid function in a GO mouse model, improves histopathological alterations, and transforms gut microbiota populations, proving its potential in treating GO through the gut-thyroid axis.
Collapse
Affiliation(s)
- Pingbo Ouyang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jia Qi
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Boding Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunping Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiamin Cao
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lujue Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tongxin Niu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Qi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
43
|
Kato T, Matsuzawa F, Shojima N, Yamauchi T. Pathogenic variants in the fibronectin type III domain of leptin receptor: Molecular dynamics simulation and structural analysis. J Mol Graph Model 2025; 135:108912. [PMID: 39608136 DOI: 10.1016/j.jmgm.2024.108912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Several case reports have identified leptin receptor (LEPR) variants associated with severe obesity in humans. However, the structure of LEPR has only been partially understood until recently, and few studies have investigated the detrimental effects of these variants on the protein's three-dimensional structure. Notably, fibronectin type III (FnIII) domains play a crucial role in signal transduction. In this study, we examined the impact of 10 variants within the FnIII domains on LEPR structure using molecular dynamics (MD) simulations and structural analysis. Our 300 ns MD simulations revealed that the C604S variant, which disrupts a key disulfide bond, significantly increased the overall root-mean-square deviation (RMSD) of the FnIII-2 and FnIII-3 domains, indicating destabilization of the interdomain rigidity required for proper signaling. Variants such as P639L, N718S, and W646C also induced abnormal bending and rotational misalignment between the FnIII domains, contributing to interdomain destabilization. Structural analysis identified folding nuclei and demonstrated that L662S, W664R, H684P, and S723F destabilize the internal domain. Variants affecting interdomain resulted in lower-than-expected damage prediction scores by bioinformatics tools. This study is expected to contribute to the elucidation of the disease-causing mechanisms of missense variants in the leptin receptor.
Collapse
Affiliation(s)
- Takashi Kato
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Fumiko Matsuzawa
- Tokyo R&D Center, Altif Laboratories, Inc., 3F Shiodome Building, 1-2-20 Kaigan, Minato-ku, Tokyo, 105-0022, Japan
| | - Nobuhiro Shojima
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
44
|
Canz MJ, Baguña-Torres J, Huerta J, Isla-Magrané H, Zufiaurre-Seijo M, Salas A, Hernandez C, Simó R, García-Arumí J, Herance JR, Bogdanov P, Duarri A. Diabetic retinopathy features in lund MetS rats. Exp Eye Res 2025; 252:110274. [PMID: 39923911 DOI: 10.1016/j.exer.2025.110274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
The Lund MetS rat (BBDR.cg-Leprdb/db.cp/LundRj) is a novel animal model that has a congenic leptin receptor deficiency (LepR-/-) and males exhibit a variety of metabolic abnormalities mimicking the human metabolic syndrome, including hyperglycemia, dyslipidemia, severe obesity, and a type 2 diabetes-like condition from 14 weeks of age. However, whether Lund MetS rats (LM rats) develop diabetic retinopathy is still unknown. The purpose is to investigate the features of diabetic retinopathy in this model. In this study, male LM rats aged 15 and 30 weeks were analyzed for pathological retinal changes, including vasculopathy, inflammation, reactive gliosis, oxidative stress, and neurodegeneration features on the retinas by histological, immunohistochemical, and gene and protein expression analysis. Compared with the non-diabetic LM rats, diabetic LM rats, mainly at 30 weeks of age, had a decrease in retinal thickness and loss of retinal ganglion cells and photoreceptors, indicating retinal neurodegeneration. They also presented an increase in VEGF-A expression, Endra, Icam-1, Vcam-1, and Endrb vascular genes, and albumin suggesting neurovascular unit dysfunction. Furthermore, retinas presented reactive gliosis and infiltration of microglia, TNF-α-positive vessels and expressed elevated levels of inflammatory genes Tnf-α, IL-18 and IL-6, and oxidative stress markers Sod2 and 8-hydroxy-2-deoxyguanosine (8-OHdG). Our results suggest that diabetic LM rats reproduce the early neurodegenerative and altered neuro-vascular features that also occur in the human diabetic eye.
Collapse
Affiliation(s)
- María José Canz
- Ophthalmology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Julia Baguña-Torres
- Medical Molecular Imaging Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Jordi Huerta
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Helena Isla-Magrané
- Ophthalmology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | | | - Anna Salas
- Ophthalmology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain
| | - Cristina Hernandez
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBER-DEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBER-DEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - José García-Arumí
- Ophthalmology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain; Departments of Medicine and Ophthalmology, Universitat Autonoma de Barcelona, Bellaterra, 08193, Spain
| | - Jose Raul Herance
- Medical Molecular Imaging Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain; CIBER-BBN (ISCIII), Instituto de Salud Carlos III (ISCIII), 28040, Madrid, Spain
| | - Patricia Bogdanov
- Medical Molecular Imaging Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBER-DEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
| | - Anna Duarri
- Ophthalmology Research Group, Vall d'Hebron Research Institute, 08035, Barcelona, Spain.
| |
Collapse
|
45
|
Semenova E, Guo A, Liang H, Hernandez CJ, John EB, Thaker VV. The expanding landscape of genetic causes of obesity. Pediatr Res 2025; 97:1358-1369. [PMID: 39690244 DOI: 10.1038/s41390-024-03780-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/18/2024] [Accepted: 11/03/2024] [Indexed: 12/19/2024]
Abstract
Obesity and weight regulation disorders are determined by the combined effects of genetics and environment. Polygenic obesity results from the combination of common variants in several genes which predisposes the individual to obesity and its related complications. In contrast, monogenic obesity results from changes in single genes, especially those in leptin-melanocortin pathway, and presents with early onset severe obesity, with or without other syndromic features. Rare variants in melanocortin 4 receptor are the commonest form of monogenic obesity. In addition, structural variation in small or large segments of chromosomes may also present with syndromic forms of obesity. Prader-Willi Syndrome, caused by imprinting errors in chromosome 15q11-13, is the most prevalent genetic cause of severe hyperphagia and obesity. With the advances in technologies, the past decade has witnessed a revolution in the identification of novel genetic causes of obesity, primarily in genes related to the leptin melanocortin pathway. The availability of safe melanocortin analogs holds the potential for targeted therapies for some of these disorders. This review summarizes known and novel rare genetic forms of obesity, along with approaches for the clinical investigation of copy number and sequence variants. The goal is to provide a reference for practicing clinicians to encourage genetic testing in obesity. IMPACT: What does this article add to the existing literature? Genetic obesity is an expanding frontier with potential to change management. Here, we summarize current information on the genetic causes of obesity and provide guidance for genetic testing. Emerging treatments may provide targeted precise treatment and change management practices.
Collapse
Affiliation(s)
- Ekaterina Semenova
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Guo
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Harry Liang
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Cindy J Hernandez
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Ella B John
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Vidhu V Thaker
- Division of Molecular Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Division of Pediatric Endocrinology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
46
|
Esbati R, Yazdani O, Simonetti J. Management of Obesity-Related Genetic Disorders. Endocrinol Metab Clin North Am 2025; 54:17-38. [PMID: 39919873 DOI: 10.1016/j.ecl.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Obesity-related genetic disorders are marked by severe, early-onset obesity caused by mutations that disrupt key biological mechanisms regulating hunger, energy balance, and fat storage. These disorders commonly impact systems such as the hypothalamic leptin-melanocortin signaling network, which plays a crucial role in controlling appetite and body weight, mainly through the melanocortin-4 receptor (MC4R) pathway. This review explores current management strategies and emerging therapies for genetic obesity disorders, highlighting the importance of treatment approaches and expanded genetic diagnostics to improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Romina Esbati
- Department of Medicine, Division of Endocrinology, Diabetes and Hypternsion, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Omid Yazdani
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical University, Boston, MA 02115, USA
| | - Juliana Simonetti
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Obesity Medicine Program, University of Utah, Salt Lake City, UT 84108, USA.
| |
Collapse
|
47
|
Vily-Petit J, Gautier-Stein A, Mithieux G. [Intestinal gluconeogenesis : When the intestine produces glucose to prevent obesity and hepatic steatosis]. Med Sci (Paris) 2025; 41:246-252. [PMID: 40117549 DOI: 10.1051/medsci/2025023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
Intestinal gluconeogenesis refers to the ability of the gut to produce glucose outside of meals. By initiating a gut-brain neural axis, its activation by dietary fiber or protein improves the regulation of energy balance. Recently, the creation of a genetic activation model of intestinal gluconeogenesis has demonstrated its anti-obesity, anti-diabetes and anti-hepatic steatosis effects. Interestingly, it increases thermogenesis in brown adipose tissue, thereby promoting energy expenditure and contributing to the fight against obesity. Therefore, targeting intestinal gluconeogenesis could be an innovative strategy to address metabolic diseases such as hepatic steatosis and diabetes, paving the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Justine Vily-Petit
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Amandine Gautier-Stein
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Gilles Mithieux
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| |
Collapse
|
48
|
Flores-Cordero JA, Aranaz-Murillo A, Vilariño-García T, Pérez-Pérez A, Izquierdo G, Flores-Campos R, Hontecillas-Prieto L, García-Domínguez DJ, Sánchez-Margalet V. Leptin and Leptin Signaling in Multiple Sclerosis: A Narrative Review. Neuromolecular Med 2025; 27:19. [PMID: 40019662 PMCID: PMC11870953 DOI: 10.1007/s12017-025-08842-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Obesity, a pandemic health problem, is now considered as a chronic inflammatory state, related to many autoimmune diseases, such as multiple sclerosis. Thus, adipokines, inflammatory mediators secreted by adipose tissue, play an important role modulating the immune response. In this context, obesity, especially during adolescent age, seems to be a key factor for the development of multiple sclerosis. Leptin, the main pro-inflammatory adipokine secreted by the adipose tissue, has been found increased in patients with multiple sclerosis and is able to regulate the immune system promoting a pro-inflammatory response. Leptin signaling in both innate and adaptative immune cells might have immunomodulatory effects in the context of multiple sclerosis. In this way, leptin has been found to produce a Th1 and Th17 response, increasing M1 macrophages and decreasing regulatory T cells and Th2 response. Moreover, circulating inflammatory adipokines, such as leptin, have been found in people with multiple sclerosis. In the present work, we are reviewing literature to update the body of knowledge regarding the role of obesity and leptin in multiple sclerosis.
Collapse
Affiliation(s)
- Juan Antonio Flores-Cordero
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
| | - Amalia Aranaz-Murillo
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, Virgen del Rocio University Hospital, Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
| | - Guillermo Izquierdo
- Neurology Service, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Rocío Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
- Department of Clinical Oncology, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío-Virgen Macarena University Hospital/CSIC/University of Seville, Seville, Spain
| | - Daniel J García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío-Virgen Macarena University Hospital/CSIC/University of Seville, Seville, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, University of Seville, Seville, Spain.
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain.
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío-Virgen Macarena University Hospital/CSIC/University of Seville, Seville, Spain.
- Department of Medical Biochemistry and Molecular Biology, and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Av. Sánchez Pizjuan 4, 41009, Seville, Spain.
| |
Collapse
|
49
|
Wu H, Li J, Jiang K, Li Y, Yu Z, Wang B, Zhou B, Zhang X, Tang N, Li Z. Characterization of Leptin and Leptin Receptor Gene in the Siberian Sturgeon ( Acipenser baerii): Molecular Cloning, Tissue Distribution, and Its Involvement in Feeding Regulation. Int J Mol Sci 2025; 26:1968. [PMID: 40076594 PMCID: PMC11900199 DOI: 10.3390/ijms26051968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Leptin is an adipokine known as a regulator of feeding and metabolism in mammals. Previous studies on fish have revealed its role in food intake regulation in limited teleosts. However, its specific function in Siberian sturgeon, an ancient Chondrostei fish, remains poorly understood. This study represents the first successful cloning of sequences for leptin and leptin receptors in Siberian sturgeon, achieved using RT-PCR. The predicted leptin sequence in this species consists of 168 amino acids that exhibit low identity with other fish species, except within the Acipenseriformes order. Tissue distribution analysis revealed a high expression of Siberian sturgeon leptin mRNA in the liver and lepr mRNA in the hypothalamus. Fasting differentially affected the expression of leptin and lepr mRNA, with decreased levels in the hypothalamus and increased levels in the liver (leptin: 3-15 days; lepr: 6-15 days). Recombinant Siberian sturgeon leptin (Ssleptin) was produced via E. coli expression, and intraperitoneal injection (100 ng/g BW) significantly inhibited food intake. The anorectic effect was correlated with changes in hypothalamic gene expression, including downregulation of orexigenic factors (agrp, orexin, npy, and ghrelin) and upregulation of anorexigenic factors (pomc, mch, and insulin). Meanwhile, the peripheral administration of Ssleptin promoted the expression of resistin in the liver and concurrently increased cck and pyy mRNA levels in the valvular intestine. Furthermore, Ssleptin injection stimulated the expression of hypothalamic lepr, jak2, akt, and ampkα2 mRNA. These findings suggest that leptin plays a significant role in the feeding control of Siberian sturgeon and provide new insights into the evolutionary function of leptin in fish.
Collapse
Affiliation(s)
- Hongwei Wu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| | - Jiamei Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| | - Kezhen Jiang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| | - Yingzi Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| | - Zhaoxiong Yu
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China
| | - Bin Wang
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China
| | - Bo Zhou
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China
| | - Xin Zhang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| | - Ni Tang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu 611731, China
| | - Zhiqiong Li
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, No. 211 Huimin Road, Chengdu 611130, China; (H.W.)
| |
Collapse
|
50
|
Dong K, Gould SI, Li M, Rivera FJS. Computational modeling of human genetic variants in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639784. [PMID: 40060429 PMCID: PMC11888284 DOI: 10.1101/2025.02.23.639784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Mouse models represent a powerful platform to study genes and variants associated with human diseases. While genome editing technologies have increased the rate and precision of model development, predicting and installing specific types of mutations in mice that mimic the native human genetic context is complicated. Computational tools can identify and align orthologous wild-type genetic sequences from different species; however, predictive modeling and engineering of equivalent mouse variants that mirror the nucleotide and/or polypeptide change effects of human variants remains challenging. Here, we present H2M (human-to-mouse), a computational pipeline to analyze human genetic variation data to systematically model and predict the functional consequences of equivalent mouse variants. We show that H2M can integrate mouse-to-human and paralog-to-paralog variant mapping analyses with precision genome editing pipelines to devise strategies tailored to model specific variants in mice. We leveraged these analyses to establish a database containing > 3 million human-mouse equivalent mutation pairs, as well as in silico-designed base and prime editing libraries to engineer 4,944 recurrent variant pairs. Using H2M, we also found that predicted pathogenicity and immunogenicity scores were highly correlated between human-mouse variant pairs, suggesting that variants with similar sequence change effects may also exhibit broad interspecies functional conservation. Overall, H2M fills a gap in the field by establishing a robust and versatile computational framework to identify and model homologous variants across species while providing key experimental resources to augment functional genetics and precision medicine applications. The H2M database (including software package and documentation) can be accessed at https://human2mouse.com.
Collapse
Affiliation(s)
- Kexin Dong
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- University of Chinese Academy of Sciences, Beijing, China
| | - Samuel I Gould
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- University of Chinese Academy of Sciences, Beijing, China
| | - Minghang Li
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Francisco J Sánchez Rivera
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|