1
|
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
|
2
|
Ahmed MB, Habib AM, Badran S, Alsherawi A, Syed A, Khoogaly H, Hammouda A, Abou-Samra AB, Doi SA. Spexin is a biomarker of the process that regulates leptin sensitivity. Peptides 2025:171416. [PMID: 40419220 DOI: 10.1016/j.peptides.2025.171416] [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: 03/16/2025] [Revised: 04/29/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
The rising prevalence of obesity poses a critical threat to global health, prompting an urgent need for a comprehensive understanding of its underlying mechanisms. This study aimed to delve into the relationship between obesity, leptin sensitivity, and gut hormones, focusing on spexin, glucagon-like peptide-1 (GLP-1), and gastric inhibitory polypeptide (GIP). We examined patients undergoing body contouring surgeries to assess how these hormones and leptin interact. Blood samples were collected at three different time points, and hormone levels were analyzed. Our findings indicated that increases in GLP-1 and decreases in GIP correlated with improved leptin sensitivity, indicated by decreased plasma leptin levels and associated with increase in steepness of the plasma leptin-spexin slope. These results suggest that spexin may serve as a biomarker for leptin sensitivity, the latter influenced by gut hormones in obese individuals. The study provides further evidence that the modulation of leptin sensitivity by gut hormones could be key in addressing obesity and its metabolic consequences.
Collapse
Affiliation(s)
- Mohamed Badie Ahmed
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar; Plastic Surgery Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Abdella M Habib
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Saif Badran
- Division of Plastic and Reconstructive Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Abeer Alsherawi
- Plastic Surgery Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar; College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Asma Syed
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hoda Khoogaly
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Atalla Hammouda
- Plastic Surgery Department, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Suhail A Doi
- Department of Population Medicine, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
3
|
Yao J, Zhu CQ, Sun Y, Huang YW, Li QH, Liao HM, Deng XJ, Li WM. Insulin resistance: The role in comorbid type 2 diabetes mellitus and depression. Neurosci Biobehav Rev 2025; 175:106218. [PMID: 40403856 DOI: 10.1016/j.neubiorev.2025.106218] [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: 01/12/2025] [Revised: 04/04/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025]
Abstract
Insulin resistance (IR) plays a significant role in the pathophysiology of comorbid type 2 diabetes mellitus (T2DM) and depression (CDD) through multifaceted mechanisms, including dysregulation of insulin signaling (both central and peripheral), neuroendocrine disturbances (hypothalamic-pituitary-adrenal axis dysfunction and monoaminergic neurotransmission impairment), chronic inflammation, oxidative stress, disruption of the microbiota-gut-brain axis, reduced brain-derived neurotrophic factor levels, and altered synaptic plasticity. These IR-related pathways may predispose individuals to depressive symptoms or exacerbate existing mood disorders. A comprehensive understanding of these mechanisms is critical for developing integrated therapeutic strategies that concurrently target metabolic and psychiatric dysfunction. Antidepressant medications exhibit divergent effects on glucose metabolism. Tricyclic antidepressants, particularly amitriptyline and nortriptyline, worsen metabolic profiles by exacerbating IR, promoting weight gain, and inducing hyperglycemia, thereby increasing diabetes risk with prolonged use. Consequently, tricyclic antidepressants should be avoided in metabolically vulnerable populations unless alternatives are unavailable. Mirtazapine presents a paradoxical profile-while its appetite-stimulating effects often lead to weight gain (a known IR risk factor), some evidence suggests potential β-cell function preservation, necessitating cautious use of mirtazapine in individuals with metabolic syndrome. Among selective serotonin reuptake inhibitors, fluoxetine and escitalopram demonstrate favorable metabolic effects, including improved insulin sensitivity and glycemic control, though hypoglycemia risk (particularly with concomitant sulfonylureas) warrants monitoring. Bupropion, a norepinephrine-dopamine reuptake inhibitor, uniquely promotes weight loss and enhances glycemic control, making it a preferred option for depression comorbid with obesity or T2DM. Agomelatine, with its neutral metabolic profile and circadian rhythm-modulating properties, represents a safer alternative for patients with metabolic concerns. Concurrently, certain antidiabetic agents show promise in managing depression. Metformin and sodium-glucose cotransporter-2 inhibitors may be prioritized for diabetic patients at risk for depression, while glucagon-like peptide-1 receptor agonists appear particularly beneficial for obesity-related mood disturbances. Thiazolidinediones offer value in treatment-resistant cases, whereas insulin secretagogues should be used cautiously in psychiatrically vulnerable individuals. Future research should prioritize three key directions: (1) Mechanistic investigations using advanced neuroimaging to elucidate the contribution of IR to depressive phenotypes and evaluate novel interventions (such as intranasal insulin); (2) Precision medicine approaches incorporating biomarkers, including genetic polymorphisms, inflammatory markers, and gut microbiome signatures, to optimize antidepressant selection and develop personalized treatment algorithms; and (3) Therapeutic innovation, including dual GLP-1/GIP agonists and anti-inflammatory-antidepressant combinations, as well as integrating digital health technologies (e.g., continuous glucose monitoring coupled with mood tracking), will enable real-time, data-driven management. These advances will be instrumental in establishing integrated care paradigms for this comorbidity, which intertwines metabolic and psychiatric conditions.
Collapse
Affiliation(s)
- Jia Yao
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Chang-Qing Zhu
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Yan Sun
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Yi-Wen Huang
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Qing-Hua Li
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Hui-Min Liao
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Xue-Jian Deng
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China
| | - Wan-Mei Li
- Department of Endocrinology, Guangzhou Twelfth People's Hospital, Guangzhou 510620, China; Department of Endocrinology, Guangzhou Occupational Disease Prevention and Treatment Hospital, Guangzhou 510620, China.
| |
Collapse
|
4
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 DOI: 10.3390/cancers17101676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
5
|
Chen LL, Tohit NM, Ludin AFM, Manaf ZA, Wong A, Kuan KL, Chien HW, Shahar S. Efficacy of diabetes-specific partial meal replacement on glycemic and weight control in type 2 diabetes: A randomized controlled trial. Diabetes Obes Metab 2025. [PMID: 40345156 DOI: 10.1111/dom.16446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/18/2025] [Accepted: 04/26/2025] [Indexed: 05/11/2025]
Abstract
AIMS To evaluate the efficacy of a diabetes-specific formula (DSF)-based partial meal replacement (PMR) in improving glycemic control, weight, and underlying hormonal changes among participants with type 2 diabetes and overweight or obesity compared to dietary consultation. MATERIALS AND METHODS This 12-week, parallel randomized controlled trial was conducted at Hospital Canselor Tuanku Muhriz, National University of Malaysia, from February 2022 to March 2024. Adults aged 20-65 years with type 2 diabetes [haemoglobin A1c (HbA1c) ≥7.5%] and overweight or obesity were randomized into two groups: PMR with DSF (Metabolic+ Sauver) plus dietary consultation (treatment group) or dietary consultation alone (control group). The primary endpoint was mean change in HbA1c at week 12. RESULTS Among 156 participants (mean age 52.2 ± 9.7 years), 141 completed the 12-week intervention. The treatment group had a greater HbA1c reduction compared to controls (-0.83% vs. -0.19%; MD: -0.63%; 95% CI: -1.00, -0.27; p < 0.001). A significantly higher proportion of the treatment group participants (61.4%) achieved clinically significant HbA1c reduction (≥0.5%) compared to controls (42.3%, p = 0.023). Fasting glucose, insulin, and anthropometric measurements also significantly improved in the treatment group compared to controls. Subsample analysis on hormonal changes revealed significant improvements in adiponectin levels among the treatment group. CONCLUSION This study demonstrated that PMR with DSF significantly improved glycemic and weight management in participants with type 2 diabetes and overweight or obesity. Adiponectin levels increased in the treatment group, correlating with improved glycemic control. No adverse events were observed on liver and kidney profiles, highlighting its potential as a safe and effective approach for diabetes management.
Collapse
Affiliation(s)
- Lew Leong Chen
- Center for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Noorlaili Mohd Tohit
- Department of Family Medicine, Faculty of Medicine, University Kebangsaan Malaysia Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | - Arimi Fitri Mat Ludin
- Center for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zahara Abdul Manaf
- Center for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Dietetic Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Alicia Wong
- Alpro Academy, Alpro Group, Sendayan Techvalley, Seremban, Malaysia
| | - Kwan Lee Kuan
- Alpro Academy, Alpro Group, Sendayan Techvalley, Seremban, Malaysia
| | - Han Wan Chien
- Alpro Academy, Alpro Group, Sendayan Techvalley, Seremban, Malaysia
| | - Suzana Shahar
- Center for Healthy Ageing and Wellness, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Dietetic Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Onu A, Trofin DM, Tutu A, Onu I, Galaction AI, Sardaru DP, Trofin D, Onita CA, Iordan DA, Matei DV. Integrative Strategies for Preventing and Managing Metabolic Syndrome: The Impact of Exercise and Diet on Oxidative Stress Reduction-A Review. Life (Basel) 2025; 15:757. [PMID: 40430185 DOI: 10.3390/life15050757] [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: 03/30/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolic syndrome (MetS) is characterized by central obesity, insulin resistance, hypertension, dyslipidemia, and chronic inflammation, significantly increasing the risk of cardiovascular disease and type 2 diabetes. Effective management of MetS is critical, with exercise being a key intervention. This review analyzed the effects of different exercise intensities-low, moderate, and high-intensity interval training (HIIT)-on metabolic health, oxidative stress (OS), inflammation, and cardiovascular function. A search of Medline, PEDro, and EBSCO identified 2251 articles, with 159 studies published between 1999 and 2025 included after screening. Low-intensity exercise improved insulin sensitivity, reduced OS markers (e.g., MDA, 8-OHdG), and enhanced antioxidant enzyme activity. Moderate-intensity exercise showed similar benefits with notable reductions in inflammatory markers (e.g., IL-1β, TNF-α). HIIT promoted fat loss and improved metabolic markers but temporarily increased OS and inflammation. Dietary strategies also play a critical role. The Mediterranean diet and Dietary Approaches to Stop Hypertension (DASH) diets are well established, emphasizing nutrient-dense foods like unsaturated fats and fiber to reduce inflammation and manage weight. The ketogenic diet (KD), a high-fat, low-carbohydrate approach, has recently gained attention for its metabolic benefits. KD induces ketosis, improving insulin sensitivity, reducing triglycerides, and enhancing fat oxidation. Studies show KD effectively reduces body weight and glucose levels, though long-term adherence and nutrient deficiencies remain challenges. Intermittent fasting also showed potential benefits, though effects on glucose metabolism were inconsistent. This review underscores the need for tailored approaches combining exercise, diet, and fasting to optimize MetS outcomes, offering integrative strategies for prevention and management.
Collapse
Affiliation(s)
- Ana Onu
- Doctoral School, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700454 Iasi, Romania
| | - Daniela-Marilena Trofin
- Doctoral School, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700454 Iasi, Romania
| | - Andrei Tutu
- Doctoral School, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700454 Iasi, Romania
| | - Ilie Onu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
| | - Anca-Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
| | - Dragos-Petrica Sardaru
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
| | - Dan Trofin
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
| | - Cristiana Amalia Onita
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
- Department of Morpho-Functional Sciences II (Pathophysiology), Center for Obesity BioBehavioral Experimental Research, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Daniel-Andrei Iordan
- Department of Individual Sports and Kinetotherapy, Faculty of Physical Education and Sport, "Dunarea de Jos" University of Galati, 800008 Galati, Romania
- Center of Physical Therapy and Rehabilitation, "Dunărea de Jos" University of Galati, 800008 Galati, Romania
| | - Daniela-Viorelia Matei
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, "Grigore T. Popa" University of Medicine and Pharmacy Iasi, 700588 Iasi, Romania
| |
Collapse
|
7
|
Ibars-Serra M, Pascual-Serrano A, Ardid-Ruiz A, Doladé N, Aguilar-González S, Cirasino J, Muguerza B, Suárez M, Keijer J, Arola-Arnal A, Aragonès G. Resveratrol Prevents Weight Gain, Counteracts Visceral Adipose Tissue Dysfunction, and Improves Hypothalamic Leptin Sensitivity in Diet-Induced Obese Rats. Mol Nutr Food Res 2025:e70075. [PMID: 40289401 DOI: 10.1002/mnfr.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
In obesity, increased adipocyte size is associated with metabolic complications, while elevated adipocyte numbers are considered a protective mechanism against metabolic disturbances. Adipose tissue dysfunction leads to decreased leptin sensitivity and disrupted energy balance regulation. Resveratrol (RSV), a bioactive compound known for potential health benefits, including obesity-related disorder prevention, has unclear modulatory effects on adipocyte dysfunction and leptin signaling in established obesity. This study investigated the impact of RSV on adiposity and hypothalamic leptin sensitivity in obesity. Rats were fed a cafeteria diet for 9 weeks and subsequently supplemented with different doses of RSV for 22 days. The 200 mg/kg RSV dose reduced leptin concentrations, body weight gain, and body fat mass in obese animals, while mitigating adipocyte hypertrophy and promoting adipocyte hyperplasia in the retroperitoneal fat depot. RSV also improved hypothalamic leptin sensitivity, shedding light on the molecular mechanisms underlying the benefits of RSV consumption for obesity-related disorders.
Collapse
Affiliation(s)
- Maria Ibars-Serra
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Aïda Pascual-Serrano
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Andrea Ardid-Ruiz
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Núria Doladé
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Aguilar-González
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Julieta Cirasino
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Begoña Muguerza
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
- Institute of Health Research Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and, Toxicological Technology (TecnATox), University Rovira i Virgili, Tarragona, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
- Institute of Health Research Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and, Toxicological Technology (TecnATox), University Rovira i Virgili, Tarragona, Spain
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Anna Arola-Arnal
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
- Institute of Health Research Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and, Toxicological Technology (TecnATox), University Rovira i Virgili, Tarragona, Spain
| | - Gerard Aragonès
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
- Institute of Health Research Pere Virgili (IISPV), Tarragona, Spain
- Center of Environmental, Food and, Toxicological Technology (TecnATox), University Rovira i Virgili, Tarragona, Spain
| |
Collapse
|
8
|
Pather R, Khathi A, Ngubane P. The effects of obesity on thyroid function in a metabolically healthy high-fat, high-carbohydrate diet-induced obese rat model. Front Endocrinol (Lausanne) 2025; 16:1538627. [PMID: 40331142 PMCID: PMC12052566 DOI: 10.3389/fendo.2025.1538627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Obesity is a recognized exacerbator of thyroid dysfunction due to its detrimental effects on energy homeostasis, appetite regulation, basal metabolic rate, thermogenesis, and metabolism. However, almost all the reported findings on obesity-related thyroid dysfunction are based on models of metabolically unhealthy obesity (MUO) in the presence of insulin resistance. There are currently no reported studies using a metabolically healthy obesity (MHO) model characterized by the absence of insulin resistance to investigate thyroid dysfunction. Hence, this study aimed to investigate the association between thyroid dysfunction and obesity in a metabolically healthy high-fat high-carbohydrate diet-induced obese rat model. Materials and methods Male Sprague Dawley rats were randomly divided into either the control diet or the high-fat high-carbohydrate diet group (HFHC) (n=9, per group). During the 5-month induction period, the control group did not develop obesity while consuming a standard diet with water. The HFHC diet group consumed the HFHC diet and water for the same duration and was diagnosed with obesity. Post-obesity confirmation, the animals continued with the respective diets for a further 7 months to maintain the obese state. Caloric intake, fasting blood glucose (FBG) and BMI were measured once a month for the duration of the experiment. Glucose homeostasis and thyroid functional parameters were assessed terminally, accompanied by satiety and pro-inflammatory markers. Results The HFHC diet group presented with higher BMI, caloric intake and FBG, and elevated insulin, HOMA-IR, Hb1Ac, leptin and IL-6 levels compared to the control diet group. The HFHC diet group presented with significantly elevated levels of TSH, fT3 and fT4. These observations suggest that thyroid homeostasis is disturbed in the obese state. However, the reported elevated glycemic status indicators and IL-6 concentrations in the HFHC diet group did not satisfy the minimum criteria to be characterized as MUO. Conclusion The HFHC diet has induced MHO in male Sprague Dawley rats. This warrants using this model to investigate the homeostatic changes that occur during the metabolically healthy obese state. This can open new avenues for developing preventative measures to avoid progressing to MUO.
Collapse
Affiliation(s)
- Reveshni Pather
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | | | | |
Collapse
|
9
|
Bertacchi V, Corley M, Aronsen GP, Bribiescas RG. Impact of high temperatures on enzyme-linked immunoassay (ELISA) performance for leptin measurements in human milk stored under varied freeze/thaw conditions. PLoS One 2025; 20:e0320366. [PMID: 40106448 PMCID: PMC11957766 DOI: 10.1371/journal.pone.0320366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Ambient temperature conditions are a common concern during laboratory analysis. Due to unexpected shipping conditions, leptin ELISA kits (Leptin Ultrasensitive, ALPCO USA; Catalog #22-LEPHUU-E01) arrived from the manufacturer at our laboratory at a temperature (76.3°F/24.6°C) well above the 2-8°C conditions recommended by the manufacturer. Since no data are available on the effects of high ambient temperature exposure on the performance of this commercial assay, we opportunistically assessed assay performance using human milk samples. Leptin measurement of recently collected and frozen human milk samples was compared between the warm temperature exposed assay kits and Normal kits that arrived and were stored at recommended temperatures (2-8 °C). We found that assay kit exposure to warm temperature during shipping resulted in sample results that were significantly different from Normal kits despite similar standard curve performance. Measurement variability from human milk samples increased with warmed kits in association with greater freeze/thaw times. This suggests that even under high temperature transportation conditions, this leptin assay performance is robust with kit reagents but compromised with human milk samples. We conclude that kits exposed to high temperature during shipment and/or storage should not be used to run human milk samples and that our concerns may extend to other biological media (i.e., serum, urine, or saliva). This study fills a critical gap in the literature on assay performance validation under non-ideal conditions, such as high temperatures. As global temperatures continue to rise, this question will become more pertinent to research integrity if left unaddressed. In light of our findings, we propose that industry standards for ELISA kit shipping and handling should be evaluated to ensure that all kits are being received in an optimal condition.
Collapse
Affiliation(s)
- Victoria Bertacchi
- Reproductive Ecology Laboratory, Department of Anthropology, Yale University, New Haven, Connecticut, United States of America
| | - Margaret Corley
- Reproductive Ecology Laboratory, Department of Anthropology, Yale University, New Haven, Connecticut, United States of America
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, United States of America
| | - Gary P. Aronsen
- Reproductive Ecology Laboratory, Department of Anthropology, Yale University, New Haven, Connecticut, United States of America
| | - Richard G. Bribiescas
- Reproductive Ecology Laboratory, Department of Anthropology, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
10
|
Zhao X, Zhang Y, Bai D, Zhen W, Guo C, Wang Z, Ma P, Ma X, Xie X, Ito K, Zhang B, Yang Y, Li J, Ma Y. Aspirin Eugenol Ester Modulates the Hypothalamus Transcriptome in Broilers Under High Stocking Density. Animals (Basel) 2025; 15:823. [PMID: 40150351 PMCID: PMC11939338 DOI: 10.3390/ani15060823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Broilers grown in a high-density (HD) stocking environment may experience intense competition that may adversely affect their growth relative to animals reared at a normal density (ND). The growth performance of HD broilers is increased by aspirin eugenol ester (AEE), although the mechanism by which this compound modulates hypothalamus-regulated feeding behavior is unclear. The aims of this study were to determine the effects of including AEE in the basal diet on the hypothalamic transcriptome and to examine in parallel the impact of these modifications on broiler production performance in HD conditions. Three hundred sixty one-day-old male Arbor Acres broilers were randomly divided into four groups: an ND group (14 broilers/m2), HD group (22 broilers/m2), ND-AEE group, and HD-AEE group. Each treatment group had 10 replicates, with 7 broilers per replicate in the ND and ND-AEE groups and 11 broilers per replicate in the HD and HD-AEE groups. Broiler growth performance was monitored, and hypothalamus samples were collected for transcriptome analysis on day 28. The HD group exhibited a reduced body weight (p < 0.01) at this timepoint compared to the ND group. However, the addition of AEE significantly improved average daily feed intake, average daily gain, and feed conversion ratio in the HD group from days 22 to 28 compared to the HD group without AEE (p < 0.05). The transcriptome results showed that 20 signaling pathways were commonly enriched among the groups (ND vs. HD, HD vs. HD-AEE). Several potential candidate genes were identified as involved in chicken central nervous system development and regulation of feed intake. Thus, the current study provides new insights into hypothalamic transcription patterns that are associated with the ameliorative effects of AEE in HD broilers.
Collapse
Affiliation(s)
- Xiaodie Zhao
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Yi Zhang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Dongying Bai
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Wenrui Zhen
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Caifang Guo
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
| | - Ziwei Wang
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
| | - Penghui Ma
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
| | - Xiqiang Ma
- Innovative Research Team of Livestock Intelligent Breeding and Equipment, Science & Technology Innovation Center for Completed Set Equipment, Longmen Laboratory, Luoyang 471023, China; (X.M.); (X.X.)
| | - Xiaolin Xie
- Innovative Research Team of Livestock Intelligent Breeding and Equipment, Science & Technology Innovation Center for Completed Set Equipment, Longmen Laboratory, Luoyang 471023, China; (X.M.); (X.X.)
| | - Koichi Ito
- Department of Food and Physiological Models, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tsukuba 319-0206, Japan;
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Yajun Yang
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Y.Y.); (J.L.)
| | - Jianyong Li
- Key Lab of New Animal Drug of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Y.Y.); (J.L.)
| | - Yanbo Ma
- Department of Animal Physiology, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, China; (X.Z.); (Y.Z.); (W.Z.); (C.G.); (Z.W.); (P.M.)
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
- Innovative Research Team of Livestock Intelligent Breeding and Equipment, Science & Technology Innovation Center for Completed Set Equipment, Longmen Laboratory, Luoyang 471023, China; (X.M.); (X.X.)
| |
Collapse
|
11
|
Yan L, Zhang X, Jin L, Li Y, Chen Y, Zhang J, Sun Z, Qi J, Qu C, Dong G, Zhang Y, Jiang Q, Liu A, Li J. The ARC CRABP1 neurons play a crucial role in the regulation of energy homeostasis. Nat Commun 2025; 16:2319. [PMID: 40057489 PMCID: PMC11890859 DOI: 10.1038/s41467-025-57411-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/19/2025] [Indexed: 05/13/2025] Open
Abstract
Recent single-cell RNA sequencing study suggested that CRABP1 expressing neurons in the arcuate nucleus (ARCCRABP1 neurons) were a distinct group of neurons. However, the physiological role of ARCCRABP1 neurons remains unexplored. Here, we demonstrated that ARCCRABP1 neurons played a crucial role in regulation of energy homeostasis in male mice. Ablation of ARCCRABP1 neurons resulted in obesity and a diabetic phenotype in mice. By employing chemogenetic or optogenetic manipulation techniques, the inhibition and activation of ARCCRABP1 neurons resulted in an increase and decrease in food intake, respectively. The axon terminals from these ARCCRABP1 neurons project to several brain regions implicated in feeding regulation such as PVH, BNST, PBN, and NTS. Optogenetic manipulation of these axons within these brain regions resulted in significant alterations of food intake behavior in mice. Furthermore, the electrophysiological studies demonstrated that the activation of ARCCRABP1 neurons induces depolarization in POMC neurons in the hypothalamus. The hormone stimulation studies showed that most of the ARCCRABP1 neurons respond to insulin. Collectively, our findings demonstrate that ARCCRABP1 neurons represent a distinct neuronal subtype involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lihong Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| | - Xin Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Liling Jin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Yin Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Yang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Jubiao Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Zhenning Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Junxia Qi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China
| | - Changqing Qu
- Fuyang Normal University, Engineering Technology Research Center of Anti-aging Chinese Herbal Medicine, Fuyang, Anhui, 236037, China
| | - Guanzhong Dong
- The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213003, China
| | - Yongjie Zhang
- Department of Human Anatomy of Nanjing Medical University, Human Brain Tissue Resource Center of Nanjing Medical University, National Health and Disease Human Brain Tissue Resource Center - sub-center of Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, 2 Sipailou Road, Nanjing, 210096, China.
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Jiangsu Provincial Key Laboratory of Molecular Targets and Intervention of Metabolic Disease, Nanjing, Jiangsu, 211166, China.
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210011, China.
| |
Collapse
|
12
|
Smith W, Azevedo EP. Hunger Games: A Modern Battle Between Stress and Appetite. J Neurochem 2025; 169:e70006. [PMID: 39936619 DOI: 10.1111/jnc.70006] [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: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Stress, an evolutionarily adaptive mechanism, has become a pervasive challenge in modern life, significantly impacting feeding-relevant circuits that play a role in the development and pathogenesis of eating disorders (EDs). Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, disrupts specific neural circuits, and dysregulates key brain regions, including the hypothalamus, hippocampus, and lateral septum. These particular structures are interconnected and key in integrating stress and feeding signals, modulating hunger, satiety, cognition, and emotional coping behaviors. Here we discuss the interplay between genetic predispositions and environmental factors that may exacerbate ED vulnerability. We also highlight the most commonly used animal models to study the mechanisms driving EDs and recent rodent studies that emphasize the discovery of novel cellular and molecular mechanisms integrating stress and feeding signals within the hippocampus-lateral septum-hypothalamus axis. In this review, we discuss the role of gut microbiome, an emerging area of research in the field of EDs and unanswered questions that persist and hinder the scientific progress, such as why some individuals remain resilient to stress while others become at high risk for the development of EDs. We finally discuss the need for future research delineating the impact of specific stressors on neural circuits, clarifying the relevance and functionality of hippocampal-septal-hypothalamic connectivity, and investigating the role of key neuropeptides such as CRH, oxytocin, and GLP-1 in human ED pathogenesis. Emerging tools like single-cell sequencing and advanced human imaging could uncover cellular and circuit-level changes in brain areas relevant for feeding in ED patients. Ultimately, by integrating basic and clinical research, science offers promising avenues for developing personalized, mechanism-based treatments targeting maladaptive eating behavior for patients suffering from EDs.
Collapse
Affiliation(s)
- Whitnei Smith
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Estefania P Azevedo
- Laboratory of Neurobiology of Behavior, Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
13
|
Ionescu VA, Gheorghe G, Bacalbasa N, Diaconu CC. Metabolic Dysfunction-Associated Steatotic Liver Disease: Pathogenetic Links to Cardiovascular Risk. Biomolecules 2025; 15:163. [PMID: 40001466 PMCID: PMC11852489 DOI: 10.3390/biom15020163] [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: 12/23/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is correlated with an increased cardiovascular risk, independent of other traditional risk factors. The mechanisms underlying this pathogenic link are complex yet remain incompletely elucidated. Among these, the most significant are visceral adiposity, low-grade inflammation and oxidative stress, endothelial dysfunction, prothrombotic status, insulin resistance, dyslipidemia and postprandial hyperlipemia, gut dysbiosis, and genetic mutations. Cardiovascular diseases are the leading cause of death in patients with MASLD. These patients have an increased incidence of coronary artery disease, carotid artery disease, structural and functional cardiac abnormalities, and valvulopathies, as well as arrhythmias and cardiac conduction disorders. In this review, we present the latest data on the association between MASLD and cardiovascular risk, focusing on the pathogenic mechanisms that explain the correlation between these two pathologies. Given the high rates of cardiovascular morbidity and mortality among patients with MASLD, we consider it imperative to raise awareness of the risks associated with this condition within the general population. Further research is essential to clarify the mechanisms underlying the increased cardiovascular risk linked to MASLD. This understanding may facilitate the identification of new diagnostic and prognostic biomarkers for these patients, as well as novel therapeutic targets.
Collapse
Affiliation(s)
- Vlad Alexandru Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Gina Gheorghe
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Nicolae Bacalbasa
- Department of Visceral Surgery, Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania;
- Department of Surgery, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania;
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
- Academy of Romanian Scientists, 050085 Bucharest, Romania
| |
Collapse
|
14
|
Oczkowicz J, Piasna-Słupecka E, Drozdowska M, Koronowicz A, Kopeć A. The Combination of Resveratrol and Conjugated Linoleic Acid Dienes Enhances the Individual Effects of These Molecules on De Novo Fatty Acid Biosynthesis in 3T3-L1 Adipocytes. Int J Mol Sci 2024; 25:13429. [PMID: 39769194 PMCID: PMC11677705 DOI: 10.3390/ijms252413429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/01/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Consuming food containing ingredients with a documented impact on lipid metabolism can help fight overweight and obesity. The simplest way to reduce the level of fatty acids is to block their synthesis or increase the rate of their degradation. This study aimed to determine the effect of resveratrol, cis-9, trans-11 conjugated linoleic acid (CLA), trans-10, cis-12 CLA, and various variants of their combinations on de novo fatty acid biosynthesis in 3T3-L1 adipocytes. The influence of the above-mentioned bioactive substances on cells grown under standard conditions and after induction of oxidative stress was measured. The effect of the tested compounds on the expression of selected genes related to the de novo fatty acid biosynthesis process (Fasn, Acc1, Acly, Prkaa1, Prkaa2, Prkaca, Srebp1) was evaluated. As part of the conducted experiments, how the level of the corresponding mRNA translates into the content of selected proteins (acetyl-CoA carboxylase 1 (ACC) and fatty acid synthase (FASN) was studied. It was found that the inhibition of fatty acid biosynthesis processes was stronger in the case of the combination of the tested CLA isomers (cis-9, trans-11 CLA, trans-10, cis-12 CLA) with resveratrol than in cases of their individual action.
Collapse
Affiliation(s)
| | | | | | | | - Aneta Kopeć
- Department of Human Nutrition and Dietetics, University of Agriculture, Balicka 122, 30-149 Kraków, Poland
| |
Collapse
|
15
|
Nagai N, Shioiri T, Hatano S, Sugiura N, Watanabe H. Regulatory role of Heparan sulfate in leptin signaling. Cell Signal 2024; 124:111456. [PMID: 39384005 DOI: 10.1016/j.cellsig.2024.111456] [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: 07/17/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Leptin, a hormone mainly secreted by adipocytes, has attracted significant attention since its discovery in 1994. Initially known for its role in appetite suppression and energy regulation, leptin is now recognized for its influence on various physiological processes, including immune response, bone formation, and reproduction. It exerts its effects by binding to receptors and initiating an intracellular signaling cascade. Heparan sulfate (HS) is known to regulate the intracellular signaling of various ligands. HS is present as the glycan portion of HSPGs on cell surfaces and in intercellular spaces, with diverse structures due to extensive sulfation and epimerization. Although HS chains on HSPGs are involved in many physiological processes, the detailed effects of HS chains on leptin signaling are not well understood. This study examined the role of HS chains on HSPGs in leptin signaling using Neuro2A cells expressing the full-length leptin receptor (LepR). We showed that cell surface HS was essential for efficient leptin signaling. Enzymatic degradation of HS significantly reduced leptin-induced phosphorylation of downstream molecules, such as signal transducer and activator of transcription 3 and p44/p42 Mitogen-activated protein kinase. In addition, HS regulated LepR expression and internalization, as treatment with HS-degrading enzymes decreased cell surface LepR. HS was also found to exhibit a weak interaction with LepR. Enzymatic removal of HS enhanced the interaction between LepR and low-density lipoprotein receptor-related protein 1, suggesting that HS negatively regulates this interaction. In conclusion, HS plays a significant role in modulating LepR availability on the cell surface, thereby influencing leptin signaling. These findings provide new insights into the complex regulation of leptin signaling and highlight potential therapeutic targets for metabolic disorders and obesity.
Collapse
Affiliation(s)
- Naoko Nagai
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Tatsumasa Shioiri
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Sonoko Hatano
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Nobuo Sugiura
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| |
Collapse
|
16
|
Huang L, Liu P, Du Y, Bazan JF, Pan D, Chen Q, Lee A, Kola VSR, Wolfe SA, Wang YX. A brown fat-enriched adipokine, ASRA, is a leptin receptor antagonist that stimulates appetite. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.12.557454. [PMID: 37745491 PMCID: PMC10515849 DOI: 10.1101/2023.09.12.557454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The endocrine control of food intake remains incompletely understood, and whether the leptin receptor (LepR)-mediated anorexigenic pathway in the hypothalamus is negatively regulated by a humoral factor is unknown. Here, we identify an appetite-stimulating factor - ASRA - that represents a peripheral signal of energy deficit and orthosterically antagonizes LepR signaling. Asra encodes an 8 kD protein that is abundantly and selectively expressed in adipose tissue and to a lesser extent, in liver. ASRA associates with autophagy vesicles and its secretion is enhanced by energy deficiency. In vivo, fasting and cold stimulate Asra expression and increase its protein concentration in cerebrospinal fluid. Asra overexpression attenuates LepR signaling, leading to elevated blood glucose and development of severe hyperphagic obesity. Conversely, either adipose- or liver-specific Asra knockout mice display increased leptin sensitivity, improved glucose homeostasis, reduced food intake, resistance to high-fat diet-induced obesity, and blunted cold-evoked feeding response. Mechanistically, ASRA acts as a high affinity antagonist of LepR. AlphaFold2-multimer prediction and mutational studies suggest that a core segment of ASRA binds to the immunoglobin-like domain of LepR, similar to the 'site 3' recognition of the A-B loop of leptin. While administration of recombinant wild-type ASRA protein promotes food intake and increases blood glucose in a LepR signaling-dependent manner, point mutation within ASRA that disrupts LepR-binding results in a loss of these effects. Our studies reveal a previously unknown endocrine mechanism in appetite regulation and have important implications for our understanding of leptin resistance.
Collapse
Affiliation(s)
- Lei Huang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - Yong Du
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- These authors contributed equally to this work: Lei Huang, Pengpeng Liu, and Yong Du
| | - J Fernando Bazan
- Bioconsulting llc, Stillwater, MN, USA
- Unit of Structural Biology, VIB-UGent Center for Inflammation Research, Gent, Belgium
| | - Dongning Pan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Present address: Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qingbo Chen
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine and Division of Hematology/Oncology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yong-Xu Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
17
|
Zou R, Zhang M, Lv W, Ren J, Fan X. Role of epicardial adipose tissue in cardiac remodeling. Diabetes Res Clin Pract 2024; 217:111878. [PMID: 39366641 DOI: 10.1016/j.diabres.2024.111878] [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: 08/08/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
Epicardial adipose tissue, or epicardial fat, is a type of visceral fat located between the heart and the pericardium. Due to its anatomical proximity to the heart, EAT plays a significant role in both cardiac physiology and pathologies, including cardiac remodeling and cardiovascular diseases (CVD). However, our understanding of how EAT pathology is influenced by risk factors such as obesity and type 2 diabetes mellitus and how altered EAT can drive cardiac remodeling and CVD, remains limited. Herein, we aimed to summarize and discuss the latest findings on EAT and its role in cardiac remodeling, highlighting the outcomes of clinical and observational studies, provide mechanistic insights, and finally introduce emerging therapeutic agents and nutritional guidelines aimed at preventing these conditions.
Collapse
Affiliation(s)
- Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Miao Zhang
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, China
| | - Weihui Lv
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, China.
| |
Collapse
|
18
|
Singhal AK, Singh G, Singh SK, Karunanand B, Gunjan G, Agrawal SK. Exploring the link between leptin levels and metabolic syndrome in elderly Indian patients: Implications for family medicine and primary care practices. J Family Med Prim Care 2024; 13:3633-3638. [PMID: 39464951 PMCID: PMC11504790 DOI: 10.4103/jfmpc.jfmpc_2008_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/17/2024] [Accepted: 02/19/2024] [Indexed: 10/29/2024] Open
Abstract
Background The metabolic syndrome (MetS), according to the Adult Treatment Panel III of the National Cholesterol Education Programme, is a collection of metabolic abnormalities that includes one, two, or all three of the following traits: obesity in the abdomen, dyslipidemia, hypertension, fasting blood sugar, or insulin resistance. This study's aim was to assess the relationship between fasting serum leptin and MetS in elderly adults with T2DM in the Northern Indian population. Material and Methods The following information was collected from all the participants: (1) anthropometric data, (2) biochemical data, and (3) a lifestyle questionnaire on sociodemographic data, dietary practices, smoking, and alcohol intake to identify their risk factors for diabetes mellitus, CVD, and hypertension. Results A total of 36 older participants (56.30%) had a history of hypertension, while 29 elderly participants (44.61%) had diabetes mellitus. A total of 32 elderly participants (49.2%) had MetS, and this group had higher serum leptin (P 0.003), body weight (P = 0.019), BMI (P 0.001), waist circumference (P 0.001), CRP (P = 0.021), insulin (P = 0.001), and HOMA-IR (P = 0.003) values as well as higher percentages of females (P = 0.001), and those with type 2 diabetes mellitus (P = 0.002) and hypertension (P = 0.039) than those in the non-MetS group. Conclusion In older persons with T2DM, our study discovered a favorable correlation between serum leptin and MetS. It can act as a standalone indicator of MetS, offering a way to spot populations at risk for associated consequences and enabling early intervention.
Collapse
Affiliation(s)
- Arjun Kumar Singhal
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT, Gurugram, Haryana, India
| | - Gaurav Singh
- Department of Microbiology, ESIC Medical College and Hospital, Patna, Bihar, India
| | - Shravan Kumar Singh
- Department of Medical Health and Family Welfare, Plibhit, Uttar Pradesh, India
| | - Busi Karunanand
- Department of Biochemistry, Faculty of Medicine and Health Sciences, SGT, Gurugram, Haryana, India
| | - Gagan Gunjan
- Department of General Medicine, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Sonu K. Agrawal
- Consultant Microbiologist, Vimta Labs. Limited, New Delhi, India
| |
Collapse
|
19
|
Liu Y, Wang Y, Zhao ZD, Xie G, Zhang C, Chen R, Zhang Y. A subset of dopamine receptor-expressing neurons in the nucleus accumbens controls feeding and energy homeostasis. Nat Metab 2024; 6:1616-1631. [PMID: 39147933 PMCID: PMC11349581 DOI: 10.1038/s42255-024-01100-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024]
Abstract
Orchestrating complex behaviors, such as approaching and consuming food, is critical for survival. In addition to hypothalamus neuronal circuits, the nucleus accumbens (NAc) also controls appetite and satiety. However, specific neuronal subtypes of the NAc that are involved and how the humoral and neuronal signals coordinate to regulate feeding remain incompletely understood. Here we decipher the spatial diversity of neuron subtypes of the NAc shell (NAcSh) and define a dopamine receptor D1-expressing and Serpinb2-expressing subtype controlling food consumption in male mice. Chemogenetics and optogenetics-mediated regulation of Serpinb2+ neurons bidirectionally regulate food seeking and consumption specifically. Circuitry stimulation reveals that the NAcShSerpinb2→LHLepR projection controls refeeding and can overcome leptin-mediated feeding suppression. Furthermore, NAcSh Serpinb2+ neuron ablation reduces food intake and upregulates energy expenditure, resulting in reduced bodyweight gain. Our study reveals a neural circuit consisting of a molecularly distinct neuronal subtype that bidirectionally regulates energy homeostasis, providing a potential therapeutic target for eating disorders.
Collapse
Affiliation(s)
- Yiqiong Liu
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Ying Wang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Zheng-Dong Zhao
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Guoguang Xie
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Chao Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Renchao Chen
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
20
|
Srour N, Caron A, Michael NJ. Do POMC neurons have a sweet tooth for leptin? Special issue: Role of nutrients in nervous control of energy balance. Biochimie 2024; 223:179-187. [PMID: 36122808 DOI: 10.1016/j.biochi.2022.09.006] [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: 05/21/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
Coordinated detection of changes in metabolic state by the nervous system is fundamental for survival. Hypothalamic pro-opiomelanocortin (POMC) neurons play a critical role in integrating metabolic signals, including leptin levels. They also coordinate adaptative responses and thus represent an important relay in the regulation of energy balance. Despite a plethora of work documenting the effects of individual hormones, nutrients, and neuropeptides on POMC neurons, the importance for crosstalk and additive effects between such signaling molecules is still underexplored. The ability of the metabolic state and the concentrations of nutrients, such as glucose, to influence leptin's effects on POMC neurons appears critical for understanding the function and complexity of this regulatory network. Here, we summarize the current knowledge on the effects of leptin on POMC neuron electrical excitability and discuss factors potentially contributing to variability in these effects, with a particular focus on the mouse models that have been developed and the importance of extracellular glucose levels. This review highlights the importance of the metabolic "environment" for determining hypothalamic neuronal responsiveness to metabolic cues and for determining the fundamental effects of leptin on the activity of hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Nader Srour
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada; Montreal Diabetes Research Center, QC, Canada.
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
21
|
Walewska E, Makowczenko KG, Witek K, Laniecka E, Molcan T, Alvarez-Sanchez A, Kelsey G, Perez-Garcia V, Galvão AM. Fetal growth restriction and placental defects in obese mice are associated with impaired decidualisation: the role of increased leptin signalling modulators SOCS3 and PTPN2. Cell Mol Life Sci 2024; 81:329. [PMID: 39090270 PMCID: PMC11335253 DOI: 10.1007/s00018-024-05336-7] [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: 09/20/2023] [Revised: 04/05/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.
Collapse
Affiliation(s)
- Edyta Walewska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karol G Makowczenko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Krzysztof Witek
- Laboratory of Cell and Tissue Analysis and Imaging, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Elżbieta Laniecka
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Andrea Alvarez-Sanchez
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Vicente Perez-Garcia
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain.
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
22
|
Mondal S, Rathor R, Singh SN, Suryakumar G. miRNA and leptin signaling in metabolic diseases and at extreme environments. Pharmacol Res Perspect 2024; 12:e1248. [PMID: 39017237 PMCID: PMC11253706 DOI: 10.1002/prp2.1248] [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: 01/01/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
The burden of growing concern about the dysregulation of metabolic processes arises due to complex interplay between environment and nutrition that has great impact on genetics and epigenetics of an individual. Thereby, any abnormality at the level of food intake regulating hormones may contribute to the development of metabolic diseases in any age group due to malnutrition, overweight, changing lifestyle, and exposure to extreme environments such as heat stress (HS), cold stress, or high altitude (HA). Hormones such as leptin, adiponectin, ghrelin, and cholecystokinin regulate appetite and satiety to maintain energy homeostasis. Leptin, an adipokine and a pleiotropic hormone, play major role in regulating the food intake, energy gain and energy expenditure. Using in silico approach, we have identified the major genes (LEP, LEPR, JAK2, STAT3, NPY, POMC, IRS1, SOCS3) that play crucial role in leptin signaling pathway. Further, eight miRNAs (hsa-miR-204-5p, hsa-miR-211-5p, hsa-miR-30, hsa-miR-3163, hsa-miR-33a-3p, hsa-miR-548, hsa-miR-561-3p, hsa-miR-7856-5p) from TargetScan 8.0 database were screened out that commonly target these genes. The role of these miRNAs should be explored as they might play vital role in regulating the appetite, energy metabolism, metabolic diseases (obesity, type 2 diabetes, cardiovascular diseases, inflammation), and to combat extreme environments. The miRNAs regulating leptin signaling and appetite may be useful for developing novel therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Samrita Mondal
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Richa Rathor
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Som Nath Singh
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | | |
Collapse
|
23
|
Vijayashankar U, Ramashetty R, Rajeshekara M, Vishwanath N, Yadav AK, Prashant A, Lokeshwaraiah R. Leptin and ghrelin dynamics: unraveling their influence on food intake, energy balance, and the pathophysiology of type 2 diabetes mellitus. J Diabetes Metab Disord 2024; 23:427-440. [PMID: 38932792 PMCID: PMC11196531 DOI: 10.1007/s40200-024-01418-2] [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: 12/29/2023] [Accepted: 03/12/2024] [Indexed: 06/28/2024]
Abstract
Purpose Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by insulin resistance and impaired glucose homeostasis. In recent years, there has been growing interest in the role of hunger and satiety hormones such as ghrelin and leptin in the development and progression of T2DM. In this context, the present literature review aims to provide a comprehensive overview of the current understanding of how ghrelin and leptin influences food intake and maintain energy balance and its implications in the pathophysiology of T2DM. Methods A thorough literature search was performed using PubMed and Google Scholar to choose the studies that associated leptin and ghrelin with T2DM. Original articles and reviews were included, letters to editors and case reports were excluded. Results This narrative review article provides a comprehensive summary on mechanism of action of leptin and ghrelin, its association with obesity and T2DM, how they regulate energy and glucose homeostasis and potential therapeutic implications of leptin and ghrelin in managing T2DM. Conclusion Ghrelin, known for its appetite-stimulating effects, and leptin, a hormone involved in the regulation of energy balance, have been implicated in insulin resistance and glucose metabolism. Understanding the complexities of ghrelin and leptin interactions in the context of T2DM may offer insights into novel therapeutic strategies for this prevalent metabolic disorder. Further research is warranted to elucidate the molecular mechanisms underlying these hormone actions and to explore their clinical implications for T2DM prevention and management.
Collapse
Affiliation(s)
- Uma Vijayashankar
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Rajalakshmi Ramashetty
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Mahesh Rajeshekara
- Department of Surgical Gastroenterology, Bangalore Medical College and Research Institute, Bangalore, 560002 India
| | - Nagashree Vishwanath
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Anshu Kumar Yadav
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru-15, Mysuru, 570015 India
| | - Akila Prashant
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru-15, Mysuru, 570015 India
| | - Rajeshwari Lokeshwaraiah
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| |
Collapse
|
24
|
Perez-Leighton C, Kerr B, Scherer PE, Baudrand R, Cortés V. The interplay between leptin, glucocorticoids, and GLP1 regulates food intake and feeding behaviour. Biol Rev Camb Philos Soc 2024; 99:653-674. [PMID: 38072002 DOI: 10.1111/brv.13039] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/21/2023] [Accepted: 12/01/2023] [Indexed: 05/09/2024]
Abstract
Nutritional, endocrine, and neurological signals converge in multiple brain centres to control feeding behaviour and food intake as part of the allostatic regulation of energy balance. Among the several neuroendocrine systems involved, the leptin, glucocorticoid, and glucagon-like peptide 1 (GLP1) systems have been extensively researched. Leptin is at the top hierarchical level since its complete absence is sufficient to trigger severe hyperphagia. Glucocorticoids are key regulators of the energy balance adaptation to stress and their sustained excess leads to excessive adiposity and metabolic perturbations. GLP1 participates in metabolic adaptation to food intake, regulating insulin secretion and satiety by parallel central and peripheral signalling systems. Herein, we review the brain and peripheral targets of these three hormone systems that integrate to regulate food intake, feeding behaviour, and metabolic homeostasis. We examine the functional relationships between leptin, glucocorticoids, and GLP1 at the central and peripheral levels, including the cross-regulation of their circulating levels and their cooperative or antagonistic actions at different brain centres. The pathophysiological roles of these neuroendocrine systems in dysregulated intake are explored in the two extremes of body adiposity - obesity and lipodystrophy - and eating behaviour disorders.
Collapse
Affiliation(s)
- Claudio Perez-Leighton
- Departmento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Carmen Sylva 2444, Providencia, Santiago, Chile
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - René Baudrand
- Departmento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
- Centro Translacional de Endocrinología (CETREN), Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| | - Víctor Cortés
- Departmento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, 830024, Chile
| |
Collapse
|
25
|
Bhalla D, Dinesh S, Sharma S, Sathisha GJ. Gut-Brain Axis Modulation of Metabolic Disorders: Exploring the Intertwined Neurohumoral Pathways and Therapeutic Prospects. Neurochem Res 2024; 49:847-871. [PMID: 38244132 DOI: 10.1007/s11064-023-04084-7] [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: 08/29/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024]
Abstract
A significant rise in metabolic disorders, frequently brought on by lifestyle choices, is alarming. A wide range of preliminary studies indicates the significance of the gut-brain axis, which regulates bidirectional signaling between the gastrointestinal tract and the cognitive system, and is crucial for regulating host metabolism and cognition. Intimate connections between the brain and the gastrointestinal tract provide a network of neurohumoral transmission that can transmit in both directions. The gut-brain axis successfully establishes that the wellness of the brain is always correlated with the extent to which the gut operates. Research on the gut-brain axis has historically concentrated on how psychological health affects how well the gastrointestinal system works. The latest studies, however, revealed that the gut microbiota interacts with the brain via the gut-brain axis to control phenotypic changes in the brain and in behavior. This study addresses the significance of the gut microbiota, the role of the gut-brain axis in management of various metabolic disorders, the hormonal and neural signaling pathways and the therapeutic treatments available. Its objective is to establish the significance of the gut-brain axis in metabolic disorders accurately and examine the link between the two while evaluating the therapeutic strategies to be incorporated in the future.
Collapse
Affiliation(s)
- Diya Bhalla
- Faculty of Life and Allied Health Sciences, MS Ramaiah University of Applied Science, Bangalore, 560048, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bangalore, 560043, India.
| | - Gonchigar Jayanna Sathisha
- Department of Post Graduate Studies and Research in Biochemistry, Jnanasahyadri, Kuvempu University, Shankaraghatta, Shimoga, 577451, India
| |
Collapse
|
26
|
Manglani K, Anika NN, Patel D, Jhaveri S, Avanthika C, Sudan S, Alimohamed Z, Tiwari K. Correlation of Leptin in Patients With Type 2 Diabetes Mellitus. Cureus 2024; 16:e57667. [PMID: 38707092 PMCID: PMC11070180 DOI: 10.7759/cureus.57667] [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] [Accepted: 04/05/2024] [Indexed: 05/07/2024] Open
Abstract
The exponential increase in diabetes mellitus (DM) poses serious public health concerns. In this review, we focus on the role of leptin in type 2 DM. The peripheral actions of leptin consist of upregulating proinflammatory cytokines which play an important role in the pathogenesis of type 2 DM and insulin resistance. Moreover, leptin is known to inhibit insulin secretion and plays a significant role in insulin resistance in obesity and type 2 DM. A literature search was conducted on Medline, Cochrane, Embase, and Google Scholar for relevant articles published until December 2023. The following search strings and Medical Subject Headings (MeSH terms) were used: "Diabetes Mellitus," "Leptin," "NPY," and "Biomarker." This article aims to discuss the physiology of leptin in type 2 DM, its glucoregulatory actions, its relationship with appetite, the impact that various lifestyle modifications can have on leptin levels, and, finally, explore leptin as a potential target for various treatment strategies.
Collapse
Affiliation(s)
- Kajol Manglani
- Internal Medicine, MedStar Washington Hospital Center, Washington, USA
| | | | - Dhriti Patel
- Medicine and Surgery, B.J. Medical College and Civil Hospital, Ahmedabad, IND
| | - Sharan Jhaveri
- Medicine and Surgery, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Gujarat University, Ahmedabad, IND
| | - Chaithanya Avanthika
- Pediatrics, Icahn School of Medicine at Mount Sinai, Elmhurst Hospital Center, New York, USA
- Medicine and Surgery, Karnataka Institute of Medical Sciences, Hubballi, IND
| | - Sourav Sudan
- Internal Medicine, Government Medical College, Rajouri, Rajouri, IND
| | - Zainab Alimohamed
- Division of Research & Academic Affairs, Larkin Health System, South Miami, USA
| | - Kripa Tiwari
- Internal Medicine, Maimonides Medical Center, New York, USA
| |
Collapse
|
27
|
Bettinetti-Luque M, Trujillo-Estrada L, Garcia-Fuentes E, Andreo-Lopez J, Sanchez-Varo R, Garrido-Sánchez L, Gómez-Mediavilla Á, López MG, Garcia-Caballero M, Gutierrez A, Baglietto-Vargas D. Adipose tissue as a therapeutic target for vascular damage in Alzheimer's disease. Br J Pharmacol 2024; 181:840-878. [PMID: 37706346 DOI: 10.1111/bph.16243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
Adipose tissue has recently been recognized as an important endocrine organ that plays a crucial role in energy metabolism and in the immune response in many metabolic tissues. With this regard, emerging evidence indicates that an important crosstalk exists between the adipose tissue and the brain. However, the contribution of adipose tissue to the development of age-related diseases, including Alzheimer's disease, remains poorly defined. New studies suggest that the adipose tissue modulates brain function through a range of endogenous biologically active factors known as adipokines, which can cross the blood-brain barrier to reach the target areas in the brain or to regulate the function of the blood-brain barrier. In this review, we discuss the effects of several adipokines on the physiology of the blood-brain barrier, their contribution to the development of Alzheimer's disease and their therapeutic potential. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Miriam Bettinetti-Luque
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Laura Trujillo-Estrada
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Garcia-Fuentes
- Unidad de Gestión Clínica Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Juana Andreo-Lopez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Raquel Sanchez-Varo
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
| | - Ángela Gómez-Mediavilla
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| | - Melissa Garcia-Caballero
- Departamento de Biología Molecular y Bioquímica, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Antonia Gutierrez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David Baglietto-Vargas
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Liu Y, Zhao ZD, Xie G, Chen R, Zhang Y. A molecularly defined NAcSh D1 subtype controls feeding and energy homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.27.530275. [PMID: 36909586 PMCID: PMC10002697 DOI: 10.1101/2023.02.27.530275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Orchestrating complex behavioral states, such as approach and consumption of food, is critical for survival. In addition to hypothalamus neuronal circuits, the nucleus accumbens (NAc) also plays an important role in controlling appetite and satiety in responses to changing external stimuli. However, the specific neuronal subtypes of NAc involved as well as how the humoral and neuronal signals coordinate to regulate feeding remain incompletely understood. Here, we deciphered the spatial diversity of neuron subtypes of the NAc shell (NAcSh) and defined a dopamine receptor D1(Drd1)- and Serpinb2-expressing subtype located in NAcSh encoding food consumption. Chemogenetics- and optogenetics-mediated regulation of Serpinb2 + neurons bidirectionally regulates food seeking and consumption specifically. Circuitry stimulation revealed the NAcSh Serpinb2 →LH LepR projection controls refeeding and can overcome leptin-mediated feeding suppression. Furthermore, NAcSh Serpinb2 + neuron ablation reduces food intake and upregulates energy expenditure resulting in body weight loss. Together, our study reveals a neural circuit consisted of molecularly distinct neuronal subtype that bidirectionally regulates energy homeostasis, which can serve as a potential therapeutic target for eating disorders.
Collapse
Affiliation(s)
- Yiqiong Liu
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
| | - Zheng-dong Zhao
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
| | - Guoguang Xie
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
| | - Renchao Chen
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Harvard Stem Cell Institute, WAB-149G, 200 Longwood Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
29
|
Mendoza-Pérez S, Orta-Méndez-Y-Sánchez I, García-Gómez RS, Ordaz-Nava G, Gracia-Mora MI, Macías-Rosales L, Rico-Morales HA, Salas-Garrido G, Durán-Domínguez-de-Bazúa MDC. Stevia rebaudiana Bertoni, an American plant used as sweetener: Study of its effects on body mass control and glycemia reduction in Wistar male and female rats. PLoS One 2024; 19:e0298251. [PMID: 38412182 PMCID: PMC10898749 DOI: 10.1371/journal.pone.0298251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/18/2023] [Indexed: 02/29/2024] Open
Abstract
Stevia rebaudiana Bertoni water extracts have been used as a natural sweetener and customary medicine by the indigenous inhabitants of South America for several hundred years. This plant was sent to Europe in the 16th century and was described by Peter Jacob Esteve in Spain. Recently the food industry has started to employ S. rebaudiana as sweetener using its glycosides after purification. Advertisement claims that Stevia glycosides is good for controling body mass and reducing glycemia. This study's objective was to evaluate the effect of S. rebaudiana leaf extract on Wistar rats as animal model to prove its effectiveness on body mass control, glycemia reduction, and other biochemical parameters. Three groups were randomly formed with 24 males and 24 females: A blank group without any sweetener, a control group drinking water with 10% glucose, and the test group ingesting a 0.94% water extract of S. rebaudiana. Body mass measurements as well as food and drink consumption were daily performed. The experiment lasted 120 days after the specimens were weaned and got used to eating solid food. Euthanasia was done and blood serum was collected to evaluate the following biochemical parameters: Glucose, triglycerides, cholesterol, insulin, glucagon, leptin, ghrelin, and glucose-dependent insulinotropic peptide, GIP. Results indicated that only female rats had statistical differences in body mass gain. No relevant effects either positive or negative were found in the biochemical parameters measured. The crude extracts of S. rebaudiana did not show any relevant changes in biochemical and hormonal profiles, changes nor body mass with respect to the blank and control groups of young and healthy rats in the age range of infancy to youth. According to the results obtained, the therapeutic properties that have been associated to S. rebaudiana consumption especially for body mass control and glycemia reduction, did not occur in young and healthy male and female rats in equivalent age to infants, young children, and youths.
Collapse
Affiliation(s)
- Samuel Mendoza-Pérez
- Faculty of Chemistry, Department of Chemical Engineering, UNAM, Laboratories of Enviromental Chemical Engineering and Chemistry, Mexico City, Mexico
| | - Itzel Orta-Méndez-Y-Sánchez
- Faculty of Chemistry, Department of Chemical Engineering, UNAM, Laboratories of Enviromental Chemical Engineering and Chemistry, Mexico City, Mexico
| | - Rolando Salvador García-Gómez
- Faculty of Chemistry, Department of Chemical Engineering, UNAM, Laboratories of Enviromental Chemical Engineering and Chemistry, Mexico City, Mexico
| | - Guillermo Ordaz-Nava
- Department of Nutrition Physiology, Molecular Nutrition Area, National Institute of Medical Sciences and Nutrition "Salvador Zubirán", INCMNSZ, Mexico City, Mexico
| | - María Isabel Gracia-Mora
- Faculty of Chemistry, UNAM, Animal Experimentation Unit, UNEXA, Complex E, Circuito de la Investigación Científica s/n, Ciudad Universitaria, Mexico City, Mexico
| | - Lucía Macías-Rosales
- Faculty of Chemistry, UNAM, Animal Experimentation Unit, UNEXA, Complex E, Circuito de la Investigación Científica s/n, Ciudad Universitaria, Mexico City, Mexico
| | - Héctor A Rico-Morales
- Faculty of Chemistry, UNAM, Animal Experimentation Unit, UNEXA, Complex E, Circuito de la Investigación Científica s/n, Ciudad Universitaria, Mexico City, Mexico
| | - Gerardo Salas-Garrido
- Faculty of Veterinary Medicine & Zootechny, Department of Pathology, UNAM, Circuito de la Investigación Científica s/n, Ciudad Universitaria, Mexico City, Mexico
| | | |
Collapse
|
30
|
Yu HY, Kim KK, Baek SH, Park CI, Jeon HJ, Song AR, Park HJ, Park IB, Kang JS, Kim JM, Kim TW, Jang SM, Cha JY, Kim J. Effect of YC-1102 on the Improvement of Obesity in High-Fat Diet-Induced Obese Mice. Curr Issues Mol Biol 2024; 46:1437-1450. [PMID: 38392211 PMCID: PMC10887656 DOI: 10.3390/cimb46020093] [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: 01/18/2024] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/24/2024] Open
Abstract
Obesity is one of the major risk factors for metabolic diseases worldwide. This study examined the effects of YC-1102, an extract derived from the roots of Rosa multiflora, on 3T3-L1 preadipocytes and high-fat diet (HFD)-induced obese mice. In vivo experiments involved the oral administration of YC-1102 (100, 150, and 200 mg/kg body weight) daily to mice for eight weeks. YC-1102 was found to downregulate the expressions of PPARγ and C/EBPα during adipogenesis, inhibiting adipocyte differentiation and upregulating the expression of PGC-1α for energy metabolism to enhance mitochondrial biogenesis and fatty acid oxidation. It has been shown that daily administration of YC-1102 to mice receiving a HFD prevented an increase in body weight and the accumulation of body fat. YC-1102 administration also reduced TG, TC, and LDL cholesterol levels, as well as glucose and leptin levels, and increased adiponectin levels, thus effectively inhibiting the metabolism of lipids. YC-1102-treated mice showed significant reductions in the mRNA expression of PPARγ and C/EBPα. The levels of PGC-1α involved in energy metabolism increased significantly in the YC-1102-treated mice when compared to the HFD-treated mice. According to the findings of this study, YC-1102 has a dual mechanism that reduces transcription factors that promote the differentiation of adipocytes and increases transcription factors that promote energy consumption.
Collapse
Affiliation(s)
- Hwa-Young Yu
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju 54907, Republic of Korea
| | - Kyoung Kon Kim
- Newgen Healthcare Co., Ltd., 56 Soyanggang-ro, Chuncheon-si 24232, Republic of Korea
| | - Sin Hwa Baek
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
| | - Cho I Park
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
| | - Hye Jin Jeon
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
| | - Ae Ri Song
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
| | - Hyun-Je Park
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
| | - Il Bum Park
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
| | - Jong Soo Kang
- Yuhan Care Co., Ltd., Seoul 07335, Republic of Korea
| | - Jung Min Kim
- Newgen Healthcare Co., Ltd., 56 Soyanggang-ro, Chuncheon-si 24232, Republic of Korea
| | - Tae Woo Kim
- Newgen Healthcare Co., Ltd., 56 Soyanggang-ro, Chuncheon-si 24232, Republic of Korea
| | - Sun Min Jang
- Newgen Healthcare Co., Ltd., 56 Soyanggang-ro, Chuncheon-si 24232, Republic of Korea
| | - Joo Young Cha
- Yuhan Care Co., Ltd., Yuhan Care R&D Center, Yongin-si 17084, Republic of Korea
- Yuhan Care Co., Ltd., Yuhan Natural Product R&D Center, Andong-si 36618, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Junghyun Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju 54907, Republic of Korea
- Non-Clinical Evaluation Center Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 57907, Republic of Korea
| |
Collapse
|
31
|
Lei Y, Liang X, Sun Y, Yao T, Gong H, Chen Z, Gao Y, Wang H, Wang R, Huang Y, Yang T, Yu M, Liu L, Yi CX, Wu QF, Kong X, Xu X, Liu S, Zhang Z, Liu T. Region-specific transcriptomic responses to obesity and diabetes in macaque hypothalamus. Cell Metab 2024; 36:438-453.e6. [PMID: 38325338 DOI: 10.1016/j.cmet.2024.01.003] [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: 06/08/2023] [Revised: 10/27/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
The hypothalamus plays a crucial role in the progression of obesity and diabetes; however, its structural complexity and cellular heterogeneity impede targeted treatments. Here, we profiled the single-cell and spatial transcriptome of the hypothalamus in obese and sporadic type 2 diabetic macaques, revealing primate-specific distributions of clusters and genes as well as spatial region, cell-type-, and gene-feature-specific changes. The infundibular (INF) and paraventricular nuclei (PVN) are most susceptible to metabolic disruption, with the PVN being more sensitive to diabetes. In the INF, obesity results in reduced synaptic plasticity and energy sensing capability, whereas diabetes involves molecular reprogramming associated with impaired tanycytic barriers, activated microglia, and neuronal inflammatory response. In the PVN, cellular metabolism and neural activity are suppressed in diabetic macaques. Spatial transcriptomic data reveal microglia's preference for the parenchyma over the third ventricle in diabetes. Our findings provide a comprehensive view of molecular changes associated with obesity and diabetes.
Collapse
Affiliation(s)
- Ying Lei
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Xian Liang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yunong Sun
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Ting Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710063, China
| | - Hongyu Gong
- School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanqing Gao
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hui Wang
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yunqi Huang
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Tao Yang
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Miao Yu
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Longqi Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingxing Kong
- School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Xun Xu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Shiping Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Zhi Zhang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China; School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China.
| |
Collapse
|
32
|
Sato S. Adipo-oncology: adipocyte-derived factors govern engraftment, survival, and progression of metastatic cancers. Cell Commun Signal 2024; 22:52. [PMID: 38238841 PMCID: PMC10797898 DOI: 10.1186/s12964-024-01474-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024] Open
Abstract
Conventional therapies for metastatic cancers have limited efficacy. Recently, cancer therapies targeting noncancerous cells in tumor microenvironments have shown improved clinical outcomes in patients. However, further advances in our understanding of the metastatic tumor microenvironment are required to improve treatment outcomes. Adipocytes are distributed throughout the body, and as a part of the metastatic tumor microenvironment, they interact with cancer cells in almost all organs. Adipocytes secrete various factors that are reported to exert clinical effects on cancer progression, including engraftment, survival, and expansion at the metastatic sites. However, only a few studies have comprehensively examined their impact on cancer cells. In this review, we examined the impact of adipocytes on cancer by describing the adipocyte-secreted factors that are involved in controlling metastatic cancer, focusing on adipokines, such as adiponectin, leptin, visfatin, chemerin, resistin, apelin, and omentin. Adipocyte-secreted factors promote cancer metastasis and contribute to various biological functions of cancer cells, including migration, invasion, proliferation, immune evasion, and drug resistance at the metastatic sites. We propose the establishment and expansion of "adipo-oncology" as a research field to enhance the comprehensive understanding of the role of adipocytes in metastatic cancers and the development of more robust metastatic cancer treatments.
Collapse
Affiliation(s)
- Shinya Sato
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, 2-3-2, Asahi-Ku, Yokohama, Kanagawa, 241-8515, Japan.
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2, Asahi-Ku, Yokohama, Kanagawa, 241-8515, Japan.
- Department of Pathology, Kanagawa Cancer Center Hospital, 2-3-2, Asahi-Ku, Yokohama, Kanagawa, 241-8515, Japan.
| |
Collapse
|
33
|
Liu S, Song S, Wang S, Cai T, Qin L, Wang X, Zhu G, Wang H, Yang W, Fang C, Wei Y, Zhou F, Yu Y, Lin S, Peng S, Li L. Hypothalamic FTO promotes high-fat diet-induced leptin resistance in mice through increasing CX3CL1 expression. J Nutr Biochem 2024; 123:109512. [PMID: 37907171 DOI: 10.1016/j.jnutbio.2023.109512] [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: 06/26/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/02/2023]
Abstract
Long-term consumption of a high-fat diet (HFD) disrupts energy homeostasis and leads to weight gain. The fat mass and obesity-associated (FTO) gene has been consistently identified to be associated with HFD-induced obesity. The hypothalamus is crucial for regulating energy balance, and HFD-induced hypothalamic leptin resistance contributes to obesity. FTO, an N6-methyladenosine (m6A) RNA methylation regulator, may be a key mediator of leptin resistance. However, the exact mechanisms remain unclear. Therefore, the present study aims to investigate the association between FTO and leptin resistance. After HFD or standard diet (SD) feeding in male mice for 22 weeks, m6A-sequencing and western blotting assays were used to identify target genes and assess protein level, and molecular interaction changes. CRISPR/Cas9 gene knockout system was employed to investigate the potential function of FTO in leptin resistance and obesity. Our data showed that chemokine (C-X3-C motif) ligand 1 (CX3CL1) was a direct downstream target of FTO-mediated m6A modification. Furthermore, upregulation of FTO/CX3CL1 and suppressor of cytokine signaling 3 (SOCS3) in the hypothalamus impaired leptin-signal transducer and activator of transcription 3 signaling, resulting in leptin resistance and obesity. Compared to wild-type (WT) mice, FTO deficiency in leptin receptor-expressing neurons of the hypothalamus significantly inhibited the upregulation of CX3CL1 and SOCS3, and partially ameliorating leptin resistance under HFD conditions. Our findings reveal that FTO involved in the hypothalamic leptin resistance and provides novel insight into the function of FTO in the contribution to hypothalamic leptin resistance and obesity.
Collapse
Affiliation(s)
- Shujing Liu
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Shiyu Song
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Shuan Wang
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Tonghui Cai
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, P.R. China
| | - Lian Qin
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Xinzhuang Wang
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Guangming Zhu
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Haibo Wang
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Wenqi Yang
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Chunlu Fang
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Yuan Wei
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Fu Zhou
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China
| | - Yang Yu
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China; School of Sport and Health Sciences, Guangzhou Sport University, Guangzhou 510500, China
| | - Shaozhang Lin
- The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, P.R. China.
| | - Shuang Peng
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China; School of Sport and Health Sciences, Guangzhou Sport University, Guangzhou 510500, China.
| | - Liangming Li
- Key Laboratory of Sports Technique, Tactics and Physical Function of General Administration of Sport of China, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P.R. China; School of Sport and Health Sciences, Guangzhou Sport University, Guangzhou 510500, China.
| |
Collapse
|
34
|
Howe SL, Holdom CJ, McCombe PA, Henderson RD, Zigman JM, Ngo ST, Steyn FJ. Associations of postprandial ghrelin, liver-expressed antimicrobial peptide 2 and leptin levels with body composition, disease progression and survival in patients with amyotrophic lateral sclerosis. Eur J Neurol 2024; 31:e16052. [PMID: 37658515 PMCID: PMC10840749 DOI: 10.1111/ene.16052] [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: 06/03/2023] [Revised: 08/06/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND AND PURPOSE Loss of appetite contributes to weight loss and faster disease progression in amyotrophic lateral sclerosis (ALS). Impairment of appetite control in ALS may include altered production or action of orexigenic (i.e., ghrelin) and anorexigenic (i.e., liver-expressed antimicrobial peptide 2 [LEAP2] and leptin) hormones. We aimed to determine if postprandial circulating ghrelin levels, LEAP2 levels, LEAP2:ghrelin molar ratio and leptin levels differ in ALS patients compared to non-neurodegenerative disease controls, and whether they are associated with disease progression and body composition. METHODS In this prospective natural history study, we assessed postprandial plasma levels of ghrelin, LEAP2 and leptin in patients with ALS (cases; n = 46) and controls (controls; n = 43). For cases, measures were compared to changes in body weight, body composition and clinical outcomes. RESULTS Postprandial ghrelin level was decreased by 52% in cases compared to controls (p = 0.013). LEAP2:ghrelin molar ratio was increased by 249% (p = 0.009), suggesting greater ghrelin resistance. Patients with lower LEAP2:ghrelin tended to have better functional capacity at assessment, as inferred by the ALS Functional Rating Scale-Revised (τ = -0.179, p = 0.086). Furthermore, ghrelin and LEAP2:ghrelin molar ratio correlated with diagnostic delay (ghrelin, τ = 0.223, p = 0.029; LEAP2:ghrelin, τ = -0.213, p = 0.037). Baseline ghrelin level, LEAP2 level, LEAP2:ghrelin ratio and leptin level were, however, not predictive of change in functional capacity during follow-up. Also, patients with higher postprandial ghrelin levels (hazard ratio [HR] 1.375, p = 0.048), and lower LEAP2:ghelin ratios (HR 0.828, p = 0.051) had an increased risk of earlier death. CONCLUSIONS Reduced postprandial ghrelin levels, coupled with increased LEAP2:ghrelin molar ratios, suggests a loss of ghrelin action in patients with ALS. Given ghrelin's actions on appetite, metabolism and neuroprotection, reduced ghrelin and greater ghrelin resistance could contribute to impaired capacity to tolerate the physiological impact of disease. Comprehensive studies are needed to explain how ghrelin and LEAP2 contribute to body weight regulation and disease progression in ALS.
Collapse
Affiliation(s)
- Stephanie L. Howe
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
| | - Cory J. Holdom
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
| | - Pamela A. McCombe
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Robert D. Henderson
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Jeffrey M. Zigman
- Center for Hypothalamic Research, Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Shyuan T. Ngo
- Australian Institute for Bioengineering and NanotechnologyThe University of QueenslandBrisbaneQueenslandAustralia
- Centre for Clinical ResearchThe University of QueenslandBrisbaneQueenslandAustralia
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Frederik J. Steyn
- Department of NeurologyRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- School of Biomedical SciencesThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
35
|
Amorim MR, Wang X, Aung O, Bevans-Fonti S, Anokye-Danso F, Ribeiro C, Escobar J, Freire C, Pho H, Dergacheva O, Branco LGS, Ahima RS, Mendelowitz D, Polotsky VY. Leptin signaling in the dorsomedial hypothalamus couples breathing and metabolism in obesity. Cell Rep 2023; 42:113512. [PMID: 38039129 PMCID: PMC10804286 DOI: 10.1016/j.celrep.2023.113512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/25/2023] [Accepted: 11/14/2023] [Indexed: 12/03/2023] Open
Abstract
Mismatch between CO2 production (Vco2) and respiration underlies the pathogenesis of obesity hypoventilation. Leptin-mediated CNS pathways stimulate both metabolism and breathing, but interactions between these functions remain elusive. We hypothesized that LEPRb+ neurons of the dorsomedial hypothalamus (DMH) regulate metabolism and breathing in obesity. In diet-induced obese LeprbCre mice, chemogenetic activation of LEPRb+ DMH neurons increases minute ventilation (Ve) during sleep, the hypercapnic ventilatory response, Vco2, and Ve/Vco2, indicating that breathing is stimulated out of proportion to metabolism. The effects of chemogenetic activation are abolished by a serotonin blocker. Optogenetic stimulation of the LEPRb+ DMH neurons evokes excitatory postsynaptic currents in downstream serotonergic neurons of the dorsal raphe (DR). Administration of retrograde AAV harboring Cre-dependent caspase to the DR deletes LEPRb+ DMH neurons and abolishes metabolic and respiratory responses to leptin. These findings indicate that LEPRb+ DMH neurons match breathing to metabolism through serotonergic pathways to prevent obesity-induced hypoventilation.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA.
| | - Xin Wang
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - O Aung
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Shannon Bevans-Fonti
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA
| | | | - Caitlin Ribeiro
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Joan Escobar
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Carla Freire
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Huy Pho
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Luiz G S Branco
- University of São Paulo, Ribeirão Preto, São Paulo 14040-904, Brazil
| | - Rexford S Ahima
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Vsevolod Y Polotsky
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA; Department of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC 20037, USA; Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA.
| |
Collapse
|
36
|
Galmiche M, Déchelotte P. Rôle de l’axe microbiote-intestin-cerveau dans la dérégulation du comportement alimentaire au cours de l’obésité et de l’hyperphagie boulimique : les mécanismes. NUTR CLIN METAB 2023; 37:2S16-2S25. [DOI: 10.1016/s0985-0562(24)00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
37
|
Everett AB, Garvey WT, Fernandez JR, Habegger K, Harper LM, Battarbee AN, Martin SL, Moore BA, Fouts AE, Bahorski J, Chandler-Laney PC. Leptin resistance in children with in utero exposure to maternal obesity and gestational diabetes. Pediatr Obes 2023; 18:e13081. [PMID: 37859518 PMCID: PMC10841866 DOI: 10.1111/ijpo.13081] [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: 05/03/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Leptin resistance occurs with obesity, but it is unknown if individuals at risk for obesity develop leptin resistance prior to obesity. OBJECTIVE Investigate whether leptin resistance is independent of weight status in children at risk for obesity due to intrauterine exposure to maternal obesity or gestational diabetes mellitus (GDM). METHODS Mother-child dyads (N = 179) were grouped by maternal pregnancy weight and GDM status: (1) normal weight, no GDM; (2) overweight/obesity, no GDM; (3) overweight/obesity with GDM. Children (4-10 years) were further stratified by current body mass index (BMI) <85th or ≥85th percentile. Leptin resistance of children and mothers was calculated as fasting leptin/fat mass index. Two-way ANOVA was used to assess whether leptin concentrations and leptin resistance differed by current weight status or in utero exposure group, after adjusting for race, sex and Tanner stage. RESULTS Children with a BMI ≥85th percentile had more leptin resistance than those with a BMI <85th percentile (p < 0.001), but leptin resistance did not differ by in utero exposure. Similarly, leptin resistance in women was associated with weight status and not prior GDM. CONCLUSIONS Results suggest that leptin concentrations are associated with obesity but not risk for obesity based on in utero exposure to maternal obesity or GDM.
Collapse
Affiliation(s)
- Alysha B. Everett
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - W. Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jose R. Fernandez
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kirk Habegger
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lorie M. Harper
- Department of Women’s Health, Division of Maternal-Fetal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Ashley N. Battarbee
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samantha L. Martin
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Women’s Reproductive Health, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bethany A. Moore
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amelia E. Fouts
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessica Bahorski
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paula C. Chandler-Laney
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
38
|
Parvaneh RR, Vajdi M, Shiraz AN, Khani M, Farshbaf SE, Farhangi MA. Prognostic value of circulating macrophage inhibitory cytokine 1-growth differentiation factor 15 (MIC-1/GDF15) in obesity: Relation with vascular endothelial growth factor (VEGF) and markers of oxidative stress. Nutr Health 2023; 29:707-713. [PMID: 35549472 DOI: 10.1177/02601060221099716] [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: 11/16/2022]
Abstract
Background: Macrophage inhibitory cytokine 1-Growth differentiation Factor 15 (MIC-1/GDF15) and vascular endothelial growth factor (VEGF) are novel regulators of obesity and energy homeostasis and food intake. Aims: In the current cross-sectional study, we aimed to evaluate MIC-1 and VEGF concentrations and their association with serum lipids and biomarkers of oxidative stress in obese individuals. Methods: Fifty six obese subjects, aged between 20-50 years old, participated in the current study. Anthropometric and nutritional parameters were assessed and serum and blood concentrations of MIC-1/GDF15, VEGF, markers of oxidative stress and serum lipids were evaluated. Results: Serum VEGF was strongly associated with serum lipids and MIC-1/GDF15 concentrations while serum MIC-1/GDF15 was associated with total cholesterol (TC) and VEGF levels. In multivariate regression analysis, serum VEGF, appetite and GPX were potent determinants of MIC-1/GDF15 concentrations while VEGF was only associated with serum MIC-1/GDF15. Conclusion: The findings of the current study demonstrated the association between MIC-1/GDF15 and VEGF. Moreover, a positive association between these cytokines and serum lipids, was also observed. The results suggest that MIC-1/GDF15 and VEGF might be considered as prognostic markers in obesity-related metabolic disorders. Although further mechanistic studies are needed to better clarify the underlying mechanism.
Collapse
Affiliation(s)
- Roghayeh Rahbar Parvaneh
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahdi Vajdi
- Department of Community Nutrition, Faculty of Nutrition, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ata Nikfam Shiraz
- School of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Khani
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Ebadpour Farshbaf
- Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
39
|
Carvalho GB, Brandão-Lima PN, Payolla TB, Lucena SEF, Sarti FM, Fisberg RM, Rogero MM. Circulating MiRNAs Are Associated With Low-grade Systemic Inflammation and Leptin Levels in Older Adults. Inflammation 2023; 46:2132-2146. [PMID: 37464054 DOI: 10.1007/s10753-023-01867-6] [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: 05/10/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023]
Abstract
Inflammaging refers to the low-grade systemic inflammation that occurs with aging present in chronic non-communicable diseases. MicroRNAs (miRNAs) are potential biomarkers for these diseases in older adults. This study aimed to assess the expression of 21 circulating miRNAs and their associations with inflammatory biomarkers in older adults. This cross-sectional study was performed with 200 individuals participating in ISA-Nutrition. The systemic low-grade inflammation score (SIS) was calculated from the plasma concentration of 10 inflammatory biomarkers. Circulating miRNA expression was assessed using the Fluidigm method. Wilcoxon-Mann-Whitney test was employed to determine differences in SIS among groups distributed according to sex and presence of MetS. Spearman's correlation was used to estimate correlations among SIS, leptin levels, miRNA expression, and variables of interest. Analyses were performed using software R version 4.2.3, with a significance level of 0.05. The final sample consisted of 193 individuals with a mean age of 69.1 (SE = 0.5) years, being 64.7% individuals with metabolic syndrome (MetS). Positive correlations were observed between leptin concentration and metabolic risk factors, and leptin concentration was higher in individuals with MetS compared to those without MetS. The expression of 15 circulating miRNAs was negatively correlated with leptin concentration. GLMs showed negative associations between miRNAs (miR-15a, miR-16, miR-223, miR-363, miR-532), leptin, and/or SIS values; and only miR-21 showed positive association with SIS values. The results suggest the presence of peripheral leptin resistance associated with low-grade inflammation and plasma expression of miRNAs in older adults. These findings suggest the potential role of miRNAs as biomarkers for cardiometabolic risk.
Collapse
Affiliation(s)
- Gabrielli B Carvalho
- Department of Nutrition, School of Public Health, University of São Paulo, 715 Dr. Arnaldo Avenue, São Paulo, SP, 01246-904, Brazil
| | - Paula N Brandão-Lima
- Department of Nutrition, School of Public Health, University of São Paulo, 715 Dr. Arnaldo Avenue, São Paulo, SP, 01246-904, Brazil
| | - Tanyara B Payolla
- Department of Nutrition, School of Public Health, University of São Paulo, 715 Dr. Arnaldo Avenue, São Paulo, SP, 01246-904, Brazil
| | - Sadraque E F Lucena
- Department of Statistics and Actuarial Sciences, Federal University of Sergipe, Marechal Rondon Avenue, São Cristóvão, SE, 49100-000, Brazil
| | - Flávia M Sarti
- School of Arts, Sciences and Humanities, University of São Paulo, 1000 Arlindo Bettio Avenue, São Paulo, SP, 03828-000, Brazil
| | - Regina M Fisberg
- Department of Nutrition, School of Public Health, University of São Paulo, 715 Dr. Arnaldo Avenue, São Paulo, SP, 01246-904, Brazil
| | - Marcelo M Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, 715 Dr. Arnaldo Avenue, São Paulo, SP, 01246-904, Brazil.
| |
Collapse
|
40
|
Lu CW, Yang KC, Chi YC, Wu TY, Chiang CH, Chang HH, Huang KC, Yang WS. Adiponectin-leptin ratio for the early detection of lean non-alcoholic fatty liver disease independent of insulin resistance. Ann Med 2023; 55:634-642. [PMID: 36790383 PMCID: PMC9937001 DOI: 10.1080/07853890.2023.2179106] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Lean Non-alcoholic Fatty Liver Disease (NAFLD) shares a similar disease burden to those of their overweight counterparts and should be detected early. We hypothesized that the adiponectin-leptin ratio (AL ratio) could be a good marker for early detection of lean NAFLD independent of insulin resistance. MATERIALS AND METHODS A total of 575 adults without diabetes were enrolled in a community-based study. The subjects were stratified into the lean controls, lean NAFLD, simple overweight/obesity and overweight/obesity NAFLD groups according to body mass index (BMI) and ultrasonographic fatty liver indicators. Serum adiponectin and leptin levels were measured by enzyme-linked immunosorbent assay. Multivariate logistic regression analyses were performed to estimate the odds ratio of having NAFLD in relation to the tertiles of serum AL concentration after adjustment. Receiver operating characteristic (ROC) analyses were applied to evaluate the diagnostic performance of the AL ratio for NAFLD. RESULTS The mean age of the participants was 42.8 ± 11.5 years. Comparing with the lean controls, the odds of having lean NAFLD for the highest versus the lowest tertile of AL ratio was 0.28(95%CI: 0.12-0.69) after adjustment. Putting AL ratio, BMI, triglyceride, AST/ALT ratio to the diagnosis performance of NAFLD, the ROC was 0.85 (95% CI: 0.82-0.88), 0.83 (95% CI 0.78-0.87) and 0.86 (95% CI 081-0.91) for all NAFLD, NAFLD in women and NAFLD in men, respectively. (p < .001). CONCLUSIONS The study revealed that the AL ratio could be a good biomarker to early distinguish lean NAFLD patients from lean controls independent of insulin resistance. [AQ3]Key messagesThe prevalence of non-alcoholic fatty liver disease (NAFLD) increases globally and is related to liver diseases and metabolic dysfunctions. Lean subset of NAFLD shares a similar disease burden to those of their overweight counterparts and should be detected early.Adiponectin-leptin ratio were associated with the severity of steatosis and was a predictor of obese NAFLD better than each single adipokine. To date, there is no investigation that explores specifically for the relationship between lean NAFLD and AL ratio.Our study found that adiponectin-leptin ratio is a sole independent marker regardless of insulin resistance in lean NAFLD. Having lean NAFLD for the highest versus the lowest tertile of adiponectin-leptin ratio was 0.28(95%CI: 0.12-0.69) after adjustment of age, sex, current smoking, exercise habits, HOMA-IR and AST/ALT. ROC for the NAFLD performance is good for the early detection (0.85; 95% CI: 0.82-0.88). Further rigorous investigation is necessary and should be promptly performed.
Collapse
Affiliation(s)
- Chia-Wen Lu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuen-Cheh Yang
- Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chiao Chi
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsan-Yu Wu
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Hsieh Chiang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hao-Hsiang Chang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chin Huang
- Department of Family Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Family Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Family Medicine, National Taiwan University Hospital, Hsin-Chu, Taiwan
| | - Wei-Shiung Yang
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
41
|
Umbayev B, Saliev T, Safarova (Yantsen) Y, Yermekova A, Olzhayev F, Bulanin D, Tsoy A, Askarova S. The Role of Cdc42 in the Insulin and Leptin Pathways Contributing to the Development of Age-Related Obesity. Nutrients 2023; 15:4964. [PMID: 38068822 PMCID: PMC10707920 DOI: 10.3390/nu15234964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
Age-related obesity significantly increases the risk of chronic diseases such as type 2 diabetes, cardiovascular diseases, hypertension, and certain cancers. The insulin-leptin axis is crucial in understanding metabolic disturbances associated with age-related obesity. Rho GTPase Cdc42 is a member of the Rho family of GTPases that participates in many cellular processes including, but not limited to, regulation of actin cytoskeleton, vesicle trafficking, cell polarity, morphology, proliferation, motility, and migration. Cdc42 functions as an integral part of regulating insulin secretion and aging. Some novel roles for Cdc42 have also been recently identified in maintaining glucose metabolism, where Cdc42 is involved in controlling blood glucose levels in metabolically active tissues, including skeletal muscle, adipose tissue, pancreas, etc., which puts this protein in line with other critical regulators of glucose metabolism. Importantly, Cdc42 plays a vital role in cellular processes associated with the insulin and leptin signaling pathways, which are integral elements involved in obesity development if misregulated. Additionally, a change in Cdc42 activity may affect senescence, thus contributing to disorders associated with aging. This review explores the complex relationships among age-associated obesity, the insulin-leptin axis, and the Cdc42 signaling pathway. This article sheds light on the vast molecular web that supports metabolic dysregulation in aging people. In addition, it also discusses the potential therapeutic implications of the Cdc42 pathway to mitigate obesity since some new data suggest that inhibition of Cdc42 using antidiabetic drugs or antioxidants may promote weight loss in overweight or obese patients.
Collapse
Affiliation(s)
- Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Timur Saliev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan;
| | - Yuliya Safarova (Yantsen)
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Aislu Yermekova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Farkhad Olzhayev
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Denis Bulanin
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan;
| | - Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (Y.S.); (A.Y.); (F.O.); (A.T.); (S.A.)
| |
Collapse
|
42
|
Hekim MG, Kelestemur MM, Bulmus FG, Bilgin B, Bulut F, Gokdere E, Ozdede MR, Kelestimur H, Canpolat S, Ozcan M. Asprosin, a novel glucogenic adipokine: a potential therapeutic implication in diabetes mellitus. Arch Physiol Biochem 2023; 129:1038-1044. [PMID: 33663304 DOI: 10.1080/13813455.2021.1894178] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVES We aimed to investigate the effects of asprosin on diabetes with a focus on serum glucose, irisin, ghrelin, leptin levels and hepatic levels of triglycerides (TG), cholesterol, low-density lipoprotein (LDL). METHODS Asprosin (10 µg/kg) was administered intraperitoneally four times at 3-day intervals and then blood and hepatic parameters above mentioned were investigated in control and diabetic mice. RESULTS The administration of asprosin increased blood glucose level in healthy animals (p = .05) whereas it did not change blood glucose level in diabetic animals. In addition, while asprosin decreased irisin level and increased ghrelin level, it did not change leptin level in diabetic mice. Therewithal, asprosin decreased the increasing levels in hepatic TG, cholesterol, and LDL in diabetic mice. CONCLUSIONS Our novel findings implicate that asprosin may be a target molecule in preventing the development and complications of diabetes.
Collapse
Affiliation(s)
| | | | - Funda Gulcu Bulmus
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Balikesir University, Balikesir, Turkey
| | - Batuhan Bilgin
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ferah Bulut
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Ebru Gokdere
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | | | - Haluk Kelestimur
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Sinan Canpolat
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - Mete Ozcan
- Department of Biophysics, Faculty of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
43
|
De Barra C, O'Shea D, Hogan AE. NK cells vs. obesity: A tale of dysfunction & redemption. Clin Immunol 2023; 255:109744. [PMID: 37604354 DOI: 10.1016/j.clim.2023.109744] [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: 06/01/2023] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023]
Abstract
Natural killer (NK) cells are critical in protecting the body against infection and cancer. NK cells can rapidly respond to these threats by directly targeting the infected or transformed cell using their cytotoxic machinery or by initiating and amplifying the immune response via their production of cytokines. Additionally, NK cells are resident across many tissues including adipose, were their role extends from host protection to tissue homeostasis. Adipose resident NK cells can control macrophage polarization via cytokine production, whilst also regulating stressed adipocyte fate using their cytotoxic machinery. Obesity is strongly associated with increased rates of cancer and a heightened susceptibility to severe infections. This is in part due to significant obesity-related immune dysregulation, including defects in both peripheral and adipose tissue NK cells. In this review, we detail the literature to date on NK cells in the setting of obesity - outlining the consequences, mechanisms and therapeutic interventions.
Collapse
Affiliation(s)
- Conor De Barra
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
| | - Donal O'Shea
- Obesity Immunology Group, Education and Research Centre, St Vincent's University Hospital, University College, Dublin 4, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland; National Children's Research Centre, Dublin 12, Ireland.
| |
Collapse
|
44
|
Zou RH, Nouraie SM, Karoleski C, Zhang Y, Sciurba FC, Forman DE, Bon J. Incident low muscle mass is associated with greater lung disease and lower circulating leptin in a tobacco-exposed longitudinal cohort. Respir Res 2023; 24:224. [PMID: 37737171 PMCID: PMC10515430 DOI: 10.1186/s12931-023-02521-3] [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: 05/16/2023] [Accepted: 08/23/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Muscle loss is prevalent in chronic obstructive pulmonary disease (COPD). Prior studies evaluating musculoskeletal dysfunction in COPD have focused on individuals with baseline low muscle mass. Currently, there is limited data evaluating clinical characteristics and outcomes associated with progression to incident low muscle mass in a tobacco-exposed cohort of individuals with baseline normal muscle mass. METHODS We evaluated 246 participants from a single-center longitudinal tobacco-exposed cohort with serial spirometry, thoracic imaging, dual energy x-ray absorptiometry (DXA) measurements, walk testing, and plasma adipokine measurements. DXA-derived fat free mass index (FFMI) and appendicular skeletal mass index (ASMI) were used as surrogates for muscle mass. Participants with incident low muscle mass (LM) at follow-up were characterized by FFMI < 18.4 kg/m2 in males and < 15.4 kg/m2 in females and/or ASMI < 7.25 kg/m2 in males and < 5.67 kg/m2 in females. RESULTS Twenty-five (10%) participants progressed to incident low muscle mass at follow-up. At baseline, the LM subgroup had greater active smoking prevalence (60% v. 38%, p = 0.04), lower FFMI (17.8 ± 1.7 kg/m2 v. 19.7 ± 2.9 kg/m2, p = 0.002), lower ASMI (7.3 ± 0.9 kg/m2 v. 8.2 ± 1.2 kg/m2, p = 0.0003), and lower plasma leptin (14.9 ± 10.1 ng/mL v. 24.0 ± 20.9 ng/mL, p = 0.04). At follow-up, the LM subgroup had higher COPD prevalence (68% v. 43%, p = 0.02), lower FEV1/FVC (0.63 ± 0.12 v. 0.69 ± 0.12, p = 0.02), lower %DLco (66.5 ± 15.9% v. 73.9 ± 16.8%, p = 0.03), and higher annual rate of FFMI decline (-0.17 kg/m2/year v. -0.04 kg/m2/year, p = 0.006). There were no differences in age, gender distribution, pack years smoking history, or walk distance. CONCLUSIONS We identified a subgroup of tobacco-exposed individuals with normal baseline muscle mass who progressed to incident DXA-derived low muscle mass. This subgroup demonstrated synchronous lung disease and persistently low circulating leptin levels. Our study suggests the importance of assessing for muscle loss in conjunction with lung function decline when evaluating individuals with tobacco exposure.
Collapse
Affiliation(s)
- Richard H Zou
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - S Mehdi Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chad Karoleski
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Frank C Sciurba
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel E Forman
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Geriatrics, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jessica Bon
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Emphysema COPD Research Center, University of Pittsburgh, Pittsburgh, PA, USA.
- Veteran Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
- UPMC Montefiore Hospital, NW628 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
45
|
Fan X, Yuan W, Huang W, Lin Z. Recent progress in leptin signaling from a structural perspective and its implications for diseases. Biochimie 2023; 212:60-75. [PMID: 37080418 DOI: 10.1016/j.biochi.2023.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/14/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
As a multi-potency cytokine, leptin not only plays a crucial role in controlling weight and energy homeostasis but also participates in the metabolic balance in the human body. Leptin is a small helical protein with a molecular weight of 16 kDa. It can interact with multiple subtypes of its receptors to initiate intracellular signal transduction and exerts physiological effects. Disturbances in leptin signaling may lead to obesity and a variety of metabolic diseases. Leptin was also found to be a critical factor in many diseases of the elderly. In this review, we focus on recent advances in the structural and molecular mechanisms of leptin signaling through its receptors with the aim of a deeper understanding of leptin-related diseases.
Collapse
Affiliation(s)
- Xiao Fan
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Wensu Yuan
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Weidong Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China.
| | - Zhi Lin
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China.
| |
Collapse
|
46
|
Xu X, Hu X, Ma G, Wang T, Wu J, Zhu X, Chen G, Zhao L, Chen J. Detecting fa leptin receptor mutation in Zucker rats with tetra-primer amplification-refractory mutation system (ARMS)-PCR. Heliyon 2023; 9:e20159. [PMID: 37809507 PMCID: PMC10559934 DOI: 10.1016/j.heliyon.2023.e20159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the genetic mutation (fa) in the gene encoding for leptin receptor, homozygous Zucker rats (fa-/-) develop excessive adiposity and become an experimental animal model in obesity and metabolic-related diseases research. Based on tetra-primer amplification refractory mutation system-polymerase chain reaction (ARMS-PCR), we developed a method to quickly genotype Zucker rats with a mutated fa allele from their wildtype littermates. The three genotypes are clearly discriminated on 2.0% agarose gel. Our method can be used as a reliable tool to set up and maintain the breeding colony in animal facilities as well as assign animals to control and treatment groups based on their genotypes for animal studies.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xinge Hu
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guodong Ma
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Tiannan Wang
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jayne Wu
- Department of Electrical Engineering and Computer Science, The University of Tennessee Knoxville, TN, 37996, United States
| | - Xiaojuan Zhu
- Office of Information Technology, The University of Tennessee Knoxville, TN, 37996, United States
| | - Guoxun Chen
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee Knoxville, TN, 37996, United States
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee Knoxville, TN, 37996, United States
| |
Collapse
|
47
|
Wei R, Li D, Jia S, Chen Y, Wang J. MC4R in Central and Peripheral Systems. Adv Biol (Weinh) 2023; 7:e2300035. [PMID: 37043700 DOI: 10.1002/adbi.202300035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/25/2023] [Indexed: 04/14/2023]
Abstract
Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.
Collapse
Affiliation(s)
- Ran Wei
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, 200240, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Sheng Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, 200025, China
| |
Collapse
|
48
|
Hu Y, Liu L, Chen Y, Zhang X, Zhou H, Hu S, Li X, Li M, Li J, Cheng S, Liu Y, Xu Y, Yan W. Cancer-cell-secreted miR-204-5p induces leptin signalling pathway in white adipose tissue to promote cancer-associated cachexia. Nat Commun 2023; 14:5179. [PMID: 37620316 PMCID: PMC10449837 DOI: 10.1038/s41467-023-40571-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer-associated cachexia is a multi-organ weight loss syndrome, especially with a wasting disorder of adipose tissue and skeletal muscle. Small extracellular vesicles (sEVs) serve as emerging messengers to connect primary tumour and metabolic organs to exert systemic regulation. However, whether and how tumour-derived sEVs regulate white adipose tissue (WAT) browning and fat loss is poorly defined. Here, we report breast cancer cell-secreted exosomal miR-204-5p induces hypoxia-inducible factor 1A (HIF1A) in WAT by targeting von Hippel-Lindau (VHL) gene. Elevated HIF1A protein induces the leptin signalling pathway and thereby enhances lipolysis in WAT. Additionally, exogenous VHL expression blocks the effect of exosomal miR-204-5p on WAT browning. Reduced plasma phosphatidyl ethanolamine level is detected in mice lack of cancer-derived miR-204-5p secretion in vivo. Collectively, our study reveals circulating miR-204-5p induces hypoxia-mediated leptin signalling pathway to promote lipolysis and WAT browning, shedding light on both preventive screenings and early intervention for cancer-associated cachexia.
Collapse
Affiliation(s)
- Yong Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China
| | - Liu Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Yong Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiaohui Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Haifeng Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Sheng Hu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xu Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Meixin Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Siyuan Cheng
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; TaiKang Center for Life and Medical Sciences; The Institute for Advanced Studies; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei, 430072, China
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430062, China.
| | - Wei Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
49
|
Xu Y, Jia X, Zhang W, Xie Q, Zhu M, Zhao Z, Hao J, Li H, Du J, Liu Y, Feng H, He J, Li H. The effects of Ascophyllum nodosum, Camellia sinensis-leaf extract, and their joint interventions on glycolipid and energy metabolism in obese mice. Front Nutr 2023; 10:1242157. [PMID: 37693249 PMCID: PMC10483828 DOI: 10.3389/fnut.2023.1242157] [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: 06/18/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023] Open
Abstract
Objectives Obesity is often associated with glucolipid and/or energy metabolism disorders. Ascophyllum nodosum extract (seaweed extract, SE) and Camellia sinensis-leaf extract (tea extract, TE) have been reported to promote positive metabolic effects through different mechanisms. We investigated the effects of SE and TE on metabolic homeostasis in diet-induced obese mice and discussed their functional characteristics. Methods Male C57BL/6J mice fed with high-fat diets for 8 weeks were established as obese models and subsequently divided into different intervention groups, followed by SE, TE, and their joint interventions for 10 weeks. Body weight and food intake were monitored. Fasting glucose and oral glucose tolerance tests were interspersed during the experiment. After the intervention, the effects on obesity control were assessed based on body composition, liver pathology section, blood lipids and glucose, respiratory exchange ratio (RER), energy expenditure (EE1, EE2, and EE3), inflammatory factors, lipid anabolism enzymes, and gut flora of the obese mice. Results After continuous gavage intervention, the mice in the intervention groups exhibited lower body weight (lower ~4.93 g, vs. HFD 38.02 g), peri-testicular fat masses (lower ~0.61 g, vs. HFD 1.92 g), and perirenal fat masses (lower ~0.21 g, vs. HFD mice 0.70 g). All interventions prevented diet-induced increases in plasma levels of glucose, adiponectin, leptin, and the inflammatory factors IL-1β and TNF-α. The RER was modified by the interventions, while the rhythm of the RER was not. Blood lipids (total cholesterol, triglycerides, and LDL) decreased and were associated with lower lipid anabolism enzymes. In addition, the SE and TE interventions altered the structure and abundance of specific flora. Different interventions inhibited the growth of different genera positively associated with obesity (Escherichia-Shigella, Helicobacter, etc.) and promoted the growth of Akkermansia and Bacteroides, thus affecting the chronic inflammatory state. Conclusion SE and TE both have synergistic effects on weight control and glucolipid metabolism regulation by improving insulin sensitivity and reducing lipid synthesis-related enzyme expression, whereas the combination of SE and TE (3:1) has a better effect on regulating energy metabolism and inhibiting chronic inflammation.
Collapse
Affiliation(s)
- Yuhan Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
- School of Public Health, Xiamen University, Xiamen, China
| | - Xiuzhen Jia
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Wei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
- School of Public Health, Xiamen University, Xiamen, China
| | - Qiaoling Xie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
- School of Public Health, Xiamen University, Xiamen, China
| | - Meizhen Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
- School of Public Health, Xiamen University, Xiamen, China
| | - Zifu Zhao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Jingyu Hao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Haoqiu Li
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Jinrui Du
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Yan Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Haotian Feng
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Jian He
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Hongwei Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, China
- School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
50
|
Cesaro A, De Michele G, Fimiani F, Acerbo V, Scherillo G, Signore G, Rotolo FP, Scialla F, Raucci G, Panico D, Gragnano F, Moscarella E, Scudiero O, Mennitti C, Calabrò P. Visceral adipose tissue and residual cardiovascular risk: a pathological link and new therapeutic options. Front Cardiovasc Med 2023; 10:1187735. [PMID: 37576108 PMCID: PMC10421666 DOI: 10.3389/fcvm.2023.1187735] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Obesity is a heterogeneous disease that affects almost one-third of the global population. A clear association has been established between obesity and cardiovascular disease (CVD). However, CVD risk is known to be related more to the local distribution of fat than to total body fat. Visceral adipose tissue (VAT) in particular has a high impact on CVD risk. This manuscript reviews the role of VAT in residual CV risk and the available therapeutic strategies for decreasing residual CV risk related to VAT accumulation. Among the many pathways involved in residual CV risk, obesity and particularly VAT accumulation play a major role by generating low-grade systemic inflammation, which in turn has a high prognostic impact on all-cause mortality and myocardial infarction. In recent years, many therapeutic approaches have been developed to reduce body weight. Orlistat was shown to reduce both weight and VAT but has low tolerability and many drug-drug interactions. Naltrexone-bupropion combination lowers body weight but has frequent side effects and is contraindicated in patients with uncontrolled hypertension. Liraglutide and semaglutide, glucagon-like peptide 1 (GLP-1) agonists, are the latest drugs approved for the treatment of obesity, and both have been shown to induce significant body weight loss. Liraglutide, semaglutide and other GLP-1 agonists also showed a positive effect on CV outcomes in diabetic patients. In addition, liraglutide showed to specifically reduce VAT and inflammatory biomarkers in obese patients without diabetes. GLP-1 agonists are promising compounds to limit inflammation in human visceral adipocytes.
Collapse
Affiliation(s)
- Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianantonio De Michele
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli “V. Monaldi”, Naples, Italy
| | - Vincenzo Acerbo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Gianmaria Scherillo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giovanni Signore
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Paolo Rotolo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Francesco Scialla
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Giuseppe Raucci
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Elisabetta Moscarella
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| | - Olga Scudiero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Ceinge Biotecnologie Avanzate Franco Salvatore S. C. a R. L., Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Cristina Mennitti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- Division of Cardiology, A.O.R.N. “Sant'Anna e San Sebastiano”, Caserta, Italy
| |
Collapse
|