1
|
Simoes MR, Bombassaro B, Gallo-Ferraz AL, Nogueira PAS, Monfort-Pires M, Zanesco AM, Valdivieso-Rivera F, Nogueira GAS, Sponton CH, Castilho RF, Velloso LA. Balb/c mice are protected from glucose and acute cold intolerance. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167725. [PMID: 40023454 DOI: 10.1016/j.bbadis.2025.167725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
The brown adipose tissue is a potential target for interventions aimed at treating obesity and other metabolic disorders. Both genetic and environmental factors are known to regulate brown adipose tissue function and exploring the interaction between these factors could unveil new mechanisms involved in the regulation of thermogenesis. In this study, we evaluated three genetically distinct mice strains submitted to two environmental factors known to modulate brown adipose tissue function, namely, cold exposure and the consumption of a high-fat diet. The comparison of Balb/c, C57BL/6, and Swiss mice revealed that Balb/c mice were the most glucose-tolerant and the most cold-tolerant. In addition, Balb/c presented the greatest brown adipose tissue oxygen consumption, which was independent of differences in uncoupling protein 1 expression and function. The search for uncoupling protein 1-independent mechanisms that could explain the greatest cold tolerance of Balb/c mice resulted in the identification of the N-acyl amino acid regulator, PM20D1, which had a greater gene expression in the brown adipose tissue of Balb/c mice as compared to the other two strains. The immunoneutralization of PM20D1 in Balb/c mice, resulted in increased blood glucose levels and worsening of cold tolerance. In addition, the in silico knockout of Pm20d1 impacted several metabolic processes, including thermogenesis, glucose tolerance, and insulin sensitivity. In conclusion, Balb/c mice are protected from glucose and acute cold intolerance, independently of the diet. We propose that PM20D1, in an uncoupling protein 1-independent fashion, can have an important role in this protection.
Collapse
Affiliation(s)
- Marcela R Simoes
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Ana Luisa Gallo-Ferraz
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Pedro A S Nogueira
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | | | - Ariane M Zanesco
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Fernando Valdivieso-Rivera
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas (UNICAMP), Campinas, São Paulo 13083-862, Brazil
| | - Guilherme A S Nogueira
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Carlos H Sponton
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas (UNICAMP), Campinas, São Paulo 13083-862, Brazil
| | - Roger F Castilho
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP 13083-888, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Campinas, São Paulo 13083-864, Brazil.
| |
Collapse
|
2
|
Pan K, Gao Y, Zong H, Zhang Y, Qi Y, Wang H, Chen W, Zhou T, Zhao J, Yin T, Guo H, Wang M, Wang H, Pang T, Zang Y, Li J. Neuronal CCL2 responds to hyperglycaemia and contributes to anxiety disorders in the context of diabetes. Nat Metab 2025; 7:1052-1072. [PMID: 40329008 DOI: 10.1038/s42255-025-01281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/17/2025] [Indexed: 05/08/2025]
Abstract
Anxiety disorders are frequently observed in patients with diabetes and can be associated with several diabetes-related factors. Here we determine that hyperglycaemia is a major cause for the development of anxiety disorders through a C-C motif chemokine ligand 2 (CCL2)-dependent mechanism. By adopting complementary strategies, we demonstrate that neuron-specific (not peripheral) CCL2 mediates anxiety-like behaviours in streptozotocin-induced diabetic mice. Mechanistically, high glucose levels induce Tonicity-responsive enhancer-binding protein (TonEBP)-dependent CCL2 expression in neurons, leading to microglial activation in a paracrine manner. Similar phenotypes are also observed in high-fat diet-induced diabetic mice, independent of insulin signalling. Furthermore, we reveal that neuronal CCL2 in the medial prefrontal cortex and ventral hippocampus synergistically induces anxiety-like behaviours, indicating brain region-specific effects on diabetic mice. Finally, we confirm that the neuronal TonEBP-CCL2 axis and inflammatory pathways are both upregulated in patients with diabetes. Conclusively, neuronal CCL2 is specifically increased by hyperglycaemia and contributes to anxiety disorders, providing additional insights into the link between diabetes and mental health disorders.
Collapse
Affiliation(s)
- Kaijun Pan
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yanan Gao
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haichao Zong
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yongmei Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yingbei Qi
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hanlin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wengang Chen
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ting Zhou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jinwen Zhao
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tao Yin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Haoran Guo
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Min Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hanmin Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tao Pang
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yi Zang
- Lingang Laboratory, Shanghai, China.
| | - Jia Li
- Metabolic Disease Research Center, Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
3
|
Zhang G, Lian Y, Li Q, Zhou S, Zhang L, Chen L, Tang J, Liu H, Li N, Pan Q, Gu Y, Lin N, Wang H, Wang X, Guo J, Zhang W, Jin Z, Xu B, Su X, Lin M, Han Q, Qin J. Vagal pathway activation links chronic stress to decline in intestinal stem cell function. Cell Stem Cell 2025; 32:778-794.e10. [PMID: 40120585 DOI: 10.1016/j.stem.2025.02.016] [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: 05/08/2024] [Revised: 01/11/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Chronic stress adversely affects intestinal health, but the specific neural pathways linking the brain to intestinal tissue are not fully understood. Here, we show that chronic stress-induced activation of the central amygdala-dorsal motor nucleus of the vagus (CeA-DMV) pathway accelerates premature aging and impairs the stemness of intestinal stem cells (ISCs). This pathway influences ISC function independently of the microbiota, the hypothalamic-pituitary-adrenal (HPA) axis, the immune response, and the sympathetic nervous system (SNS). Under chronic stress, DMV-mediated vagal activation prompts cholinergic enteric neurons to release acetylcholine (ACh), which engages ISCs via the M3 muscarinic acetylcholine receptor (CHRM3). This interaction activates the p38 mitogen-activated protein kinase (MAPK) pathway, triggering growth arrest and mitochondrial fragmentation, thereby accelerating an aging-like decline in ISCs. Together, our findings provide insights into an alternative neural mechanism that links stress to intestinal dysfunction. Strategies targeting the DMV-associated vagal pathway represent potential therapeutic approaches for stress-induced intestinal diseases.
Collapse
Affiliation(s)
- Guoying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yannan Lian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Shudi Zhou
- Department of Endocrinology, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Lili Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Liting Chen
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junzhe Tang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, Shanghai 200032, China
| | - Hailong Liu
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Qiang Pan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China
| | - Yongqiang Gu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Naiheng Lin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Hanling Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xuege Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Jiacheng Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Zige Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Beitao Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Xiao Su
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Qi Han
- Department of Emergency and Critical Disease, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China; Jinfeng Laboratory, Chongqing 401329, China.
| |
Collapse
|
4
|
Toledo M, Martínez-Martínez S, Van Hul M, Laudo B, Eyre E, Pelicaen R, Puel A, Altirriba J, Gómez-Valadés AG, Inderhees J, Moreno-Indias I, Pozo M, Chivite I, Milà-Guasch M, Haddad-Tóvolli R, Obri A, Fos-Domènech J, Tahiri I, Llana SR, Ramírez S, Monelli E, Schwaninger M, Cani PD, Nogueiras R, Claret M. Rapid modulation of gut microbiota composition by hypothalamic circuits in mice. Nat Metab 2025:10.1038/s42255-025-01280-3. [PMID: 40263603 DOI: 10.1038/s42255-025-01280-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
In recent years, the gut microbiota and derived metabolites have emerged as relevant players in modulating several brain functions, including energy balance control1-3. This form of distant communication mirrors that of metabolic hormones (for example, leptin, ghrelin), which convey information about the organism's energy status by exerting effects on diverse brain regions, including the master homeostatic centre, the hypothalamus4. However, whether the hypothalamus is also able to influence gut microbiota composition remains enigmatic. Here we present a study designed to unravel this challenging question. To this aim, we used chemogenetics5 (to selectively activate or inhibit hypothalamic pro-opiomelanocortin or agouti-related peptide neurons) or centrally administered leptin or ghrelin to male mice. Subsequently, we conducted microbiota composition analysis throughout the gut using 16S rRNA gene sequencing. Our results showed that these brain interventions significantly changed the gut microbiota in an anatomical and short-term (2-4 h) fashion. Transcriptomic analysis indicated that these changes were associated with the reconfiguration of neuronal and synaptic pathways in the duodenum concomitant with increased sympathetic tone. Interestingly, diet-induced obesity attenuated the brain-mediated changes triggered by leptin in gut microbiota communities and sympathetic activation. Our findings reveal a previously unanticipated brain-gut axis that acutely attunes microbiota composition on fast timescales, with potential implications for meal-to-meal adjustments and systemic energy balance control.
Collapse
Affiliation(s)
- Míriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Martínez-Martínez
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Berta Laudo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rudy Pelicaen
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Anthony Puel
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julica Inderhees
- Bioanalytic Core Facility, Center for Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iasim Tahiri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergio R Llana
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Erika Monelli
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Markus Schwaninger
- German Research Centre for Cardiovascular Research (DZHK), Lübeck, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Patrice D Cani
- Metabolism and Nutrition Research Group (MNUT), Louvain Drug Research Institute (LDRI), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium.
| | - Rubén Nogueiras
- Department of Physiology (CIMUS), School of Medicine-Instituto de Investigaciones Sanitarias (IDIS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.
- Galicia Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
5
|
Mravec B, Szantova M. Liver Neurobiology: Regulation of Liver Functions by the Nervous System. Semin Liver Dis 2025. [PMID: 40239709 DOI: 10.1055/a-2562-2000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The nervous system plays an important role in the regulation of liver functions during physiological as well as pathological conditions. This regulatory effect is based on the processing of signals transmitted to the brain by sensory nerves innervating the liver tissue and other visceral organs and by humoral pathways transmitting signals from peripheral tissues and organs. Based on these signals, the brain modulates metabolism, detoxification, regeneration, repair, inflammation, and other processes occurring in the liver. The nervous system thus determines the functional and morphological characteristics of the liver. Liver innervation also mediates the influence of psychosocial factors on liver functions. The aim of this review is to describe complexity of bidirectional interactions between the brain and liver and to characterize the mechanisms and pathways through which the nervous system influences liver function during physiological conditions and maintains liver and systemic homeostasis.
Collapse
Affiliation(s)
- Boris Mravec
- Department of Physiology Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
6
|
Pace LA, Kong N, Itani MI, Hemp J. The Neuroimmune Axis in Gastrointestinal Disorders - An Underrecognized Problem. Curr Gastroenterol Rep 2025; 27:28. [PMID: 40232527 DOI: 10.1007/s11894-025-00973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE OF REVIEW We present an introduction to the neuroimmune axis with a focus on the gastrointestinal system, its role in numerous chronic multisystem disorders, and emerging tools and therapies to diagnose and treat these conditions. RECENT FINDINGS There have recently been tremendous breakthroughs in our understanding of how the nervous, immune, and endocrine systems, as well as the extracellular matrix and microbiota, interact within the gastrointestinal system to modulate health and disease.
Collapse
Affiliation(s)
- Laura A Pace
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, CA, USA.
- meliora.bio, Palo Alto, CA, USA.
| | - Niwen Kong
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Mohamed I Itani
- Division of Neurology and Neurological Sciences, Department of Medicine, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
7
|
Karimian A, Shokri K, Mohammadi A, Frishman WH, Aronow WS. Treatment of Cardiovascular Manifestations in Transgender Individuals. Cardiol Rev 2025:00045415-990000000-00459. [PMID: 40167304 DOI: 10.1097/crd.0000000000000902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The transgender population has been steadily increasing, with more individuals seeking gender-affirming care to align their physical characteristics with their gender identity. Despite advances in healthcare, transgender individuals face significant barriers to accessing culturally competent care, resulting in heightened cardiovascular risks and disparities. Cardiovascular disease prevalence among transgender individuals is influenced by a combination of physiological, psychological, and social factors, including the impacts of gender-affirming hormone therapy, surgical interventions, and minority stress. This review examines the cardiovascular risks associated with estrogen therapy in transfeminine individuals and testosterone therapy in transmasculine individuals, highlighting their effects on lipid profiles, thromboembolic risks, and metabolic parameters. Furthermore, it explores the implications of gender-affirming surgeries and the role of psychosocial stress in cardiovascular outcomes. Current evidence underscores the need for tailored risk assessment, proactive management strategies, and lifestyle interventions to optimize cardiovascular health in this population. Significant research gaps remain regarding the long-term cardiovascular effects of gender-affirming care. Large-scale, prospective studies and the development of transgender-specific cardiovascular care guidelines are crucial to address these gaps. This review advocates for a multidisciplinary, patient-centered approach to mitigate cardiovascular risks and improve outcomes for transgender individuals.
Collapse
Affiliation(s)
- Azin Karimian
- From the Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kasra Shokri
- From the Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Mohammadi
- Internal Medicine Department, Valley Health System, Las Vegas, NV
| | | | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
8
|
Zhang L, Li Y, Gao W, Li Z, Wu T, Lang C, Rui L, Zhang W. Deficiency of neuronal LGR4 increases energy expenditure and inhibits food intake via hypothalamic leptin signaling. EMBO Rep 2025; 26:2098-2120. [PMID: 40069508 PMCID: PMC12018946 DOI: 10.1038/s44319-025-00398-5] [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: 06/04/2024] [Revised: 12/18/2024] [Accepted: 01/24/2025] [Indexed: 04/25/2025] Open
Abstract
The metabolic effects of leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) remain largely unknown. Here, we showed that knockdown of Lgr4 in nestin progenitor or Sp1 mature neurons reduced high fat diet (HFD)-induced obesity by increasing energy expenditure and inhibiting food intake. Deficiency of LGR4 in AgRP neurons increased energy expenditure, and inhibited food intake, leading to alterations in glucose and lipid metabolism. Knock-down of Lgr4 in Sf1 neurons enhanced energy expenditure, reduced adiposity, and improved glucose and lipid metabolism. The metabolic benefits of neuronal LGR4 occurred via improvement of leptin signaling in AgRP and Sf1 neurons. Knockdown of Lgr4 in nestin, Sp1, AgRP or Sf1 neurons decreased hypothalamic levels of SOCS-3, and increased phosphorylation of STAT3. These alterations were associated with a significant reduction in the hypothalamic levels of β-catenin. Inhibition of β-catenin signaling by Dkk1 significantly attenuated the decrement of phospho-STAT3 and concurrent increase of SOCS-3 induced by Rspondin 3, an endogenous ligand for LGR4. Our results thus demonstrate that hypothalamic LGR4 may promote energy conversation by increasing food intake and decreasing energy expenditure. Deficiency of neuronal LGR4 improves hypothalamic leptin sensitivity via suppression of β-catenin signaling.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yuan Li
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Wenbin Gao
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Ziru Li
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Tong Wu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Chunhui Lang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Liangyou Rui
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Weizhen Zhang
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
9
|
Setarehaseman A, Mohammadi A, Maitta RW. Thrombocytopenia in Sepsis. Life (Basel) 2025; 15:274. [PMID: 40003683 PMCID: PMC11857489 DOI: 10.3390/life15020274] [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: 12/31/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Platelets, traditionally known for their role in hemostasis, have emerged as key players in immune response and inflammation. Sepsis, a life-threatening condition characterized by systemic inflammation, often presents with thrombocytopenia, which at times, can be significant. Platelets contribute to the inflammatory response by interacting with leukocytes, endothelial cells, and the innate immune system. However, excessive platelet activation and consumption can lead to thrombocytopenia and exacerbate the severity of sepsis. Understanding the multifaceted roles of platelets in sepsis is crucial for developing effective therapeutic strategies. Targeting platelet-mediated inflammatory responses and promoting platelet production may offer potential avenues for improving outcomes in septic patients with thrombocytopenia. Future research should focus on elucidating the mechanisms underlying platelet dysfunction in sepsis and exploring novel therapeutic approaches to optimize platelet function and mitigate inflammation. This review explores the intricate relationship between platelets, inflammation, and thrombosis in the context of sepsis.
Collapse
Affiliation(s)
- Alireza Setarehaseman
- University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Abbas Mohammadi
- Department of Internal Medicine, Valley Health System, Las Vegas, NV 89119, USA;
| | - Robert W. Maitta
- University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| |
Collapse
|
10
|
Jiang L, Cai S, Weng Z, Zhang S, Jiang SH. Peripheral, central, and chemotherapy-induced neuropathic changes in pancreatic cancer. Trends Neurosci 2025; 48:124-139. [PMID: 39730257 DOI: 10.1016/j.tins.2024.11.008] [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: 08/21/2024] [Revised: 10/27/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024]
Abstract
In pancreatic cancer, significant alterations occur in the local nervous system, including axonogenesis, neural remodeling, perineural invasion, and perineural neuritis. Pancreatic cancer can impact the central nervous system (CNS) through cancer cell-intrinsic factors or systemic factors, particularly in the context of cancer cachexia. These peripheral and central neuropathic changes exert substantial influence on cancer initiation and progression. Moreover, chemotherapy-induced neuropathy is common in pancreatic cancer, causing peripheral nerve damage and cognitive dysfunction. Targeting the crosstalk between pancreatic cancer and the nervous system, either peripherally or centrally, holds promise in cancer treatment, pain relief, and improved quality of life. Here, we summarize recent findings on the molecular mechanisms behind these neuropathic changes in pancreatic cancer and discuss potential intervention strategies.
Collapse
Affiliation(s)
- Luju Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shuqi Cai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Zheqi Weng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
11
|
Rauchenwald T, Benedikt-Kühnast P, Eder S, Grabner GF, Forstreiter S, Lang M, Sango R, Eisenberg T, Rattei T, Haschemi A, Wolinski H, Schweiger M. Clearing the path for whole-mount labeling and quantification of neuron and vessel density in adipose tissue. J Cell Sci 2025; 138:JCS263438. [PMID: 39878039 PMCID: PMC11832183 DOI: 10.1242/jcs.263438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/29/2024] [Indexed: 01/31/2025] Open
Abstract
White adipose tissue (WAT) comprises a plethora of cell types beyond adipocytes forming a regulatory network that ensures systemic energy homeostasis. Intertissue communication is facilitated by metabolites and signaling molecules that are spread by vasculature and nerves. Previous works have indicated that WAT responds to environmental cues by adapting the abundance of these 'communication routes'; however, the high intra-tissue heterogeneity questions the informative value of bulk or single-cell analyses and underscores the necessity of whole-mount imaging. The applicability of whole-mount WAT-imaging is currently limited by two factors - (1) methanol-based tissue clearing protocols restrict the usable antibody portfolio to methanol-resistant antibodies and (2) the vast amounts of data resulting from 3D imaging of whole-tissue samples require high computational expertise and advanced equipment. Here, we present a protocol for whole-mount WAT clearing, overcoming the constraints of antibody-methanol sensitivity. Additionally, we introduce TiNeQuant (for 'tissue network quantifier') a Fiji tool for automated 3D quantification of neuron or vascular network density, which we have made freely available. Given TiNeQuants versatility beyond WAT, it simplifies future efforts studying neuronal or vascular alterations in numerous pathologies.
Collapse
Affiliation(s)
- Thomas Rauchenwald
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Pia Benedikt-Kühnast
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Sandra Eder
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Gernot F. Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria
| | | | - Michaela Lang
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Roko Sango
- Centre for Pathobiochemistry and Genetics, Medical University of Vienna, 1090 Vienna, Austria
- Centre for Microbiology and Environmental Systems’ Science, University of Vienna, 1030 Vienna, Austria
- Doctoral School in Microbiology and Environmental Science, University of Vienna, 1030 Vienna, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
- Field of Excellence BioHealth - University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems’ Science, University of Vienna, 1030 Vienna, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine (KILM), Medical University of Vienna, 1090 Vienna, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
- Field of Excellence BioHealth - University of Graz, 8010 Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
- Field of Excellence BioHealth - University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
12
|
López M, Gualillo O. Rheumatic diseases and metabolism: where centre and periphery meet. Nat Rev Rheumatol 2024; 20:783-794. [PMID: 39478099 DOI: 10.1038/s41584-024-01178-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/26/2024]
Abstract
Over the past few decades, the connection between metabolism and various inflammatory and rheumatic diseases has been an area of active investigation. Nonetheless, the precise mechanisms underlying these relationships remain a topic of ongoing debate, owing in part to conflicting data. This discrepancy can be attributed to the predominant focus on peripheral mechanisms in research into the metabolic consequences of rheumatic diseases. However, a wealth of evidence supports the notion that the central nervous system, specifically the hypothalamus, has an important influence on metabolic homeostasis. Notably, links have been established between crucial hypothalamic mechanisms responsible for regulating energy balance (including food intake, thermogenesis, and glucose and lipid metabolism), such as AMP-activated protein kinase, and the pathophysiology of rheumatoid arthritis. This Review aims to comprehensively examine the current understanding of central metabolic control in rheumatic diseases and explore potential therapeutic options that target this pathophysiological mechanism.
Collapse
Affiliation(s)
- Miguel López
- NeurObesity Group, Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Santiago de Compostela, Spain.
| | - Oreste Gualillo
- Servizo Galego de Saude (SERGAS)-Instituto de Investigación Sanitaria de Santiago (IDIS), the Neuroendocrine Interactions in Rheumatology and Inflammatory Disease (NEIRID) Lab, Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Wei Y, Lin D, Lian Y, Wei Q, Zheng L, Yuan K, Zhao J, Kuang K, Tang Y, Gao Y. Population data study reveals pain as a possible protective factor against cerebrovascular disease in cancer patients. Sci Rep 2024; 14:29471. [PMID: 39604497 PMCID: PMC11603069 DOI: 10.1038/s41598-024-80668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
The purpose of this study is to investigate the relationship between chronic pain and the mortality rate of cerebrovascular disease (CVD) in cancer patients. Thus, we performed a case-control investigation was conducted by utilizing data from the Surveillance, Epidemiology, and End Results (SEER) database between 1975 and 2019. Multiple demographics, pain rating and other clinical characteristics were extracted to assess predictors for the death from CVD in cancer patients. Different machine learning algorithms were applied to construct pain-related prediction model. The analysis involved 16,850 case patients and 710,729 controls. Among cancer patients, approximately 2.3% succumbed to subsequent CVD. Cancer pain (Pain rating II) was associated with a decreased risk of CVD. Univariate and multivariate COX analyses indicated that older age at cancer diagnosis, male gender, single marital status, Black or Other race, and lack of systemic therapy correlated with a higher risk of CVD-related death. Propensity score matching revealed a significantly lower proportion of Pain rating II in the case group. The logistic regression algorithm demonstrated superior predictive ability for 5-year and 10-year CVD risk in cancer patients. Notably, survival time, age, and pain rating emerged as the top three crucial variables. This study firstly investigated pain and various risk factors for CVD in cancer patients, highlighting pain as a novel and possible protective factor for CVD. The development of a risk model based on pain could aid in identifying individuals at high risk for CVD and may inspire innovative strategies for preventing CVD in cancer patients.
Collapse
Affiliation(s)
- Yongbao Wei
- Shengli Clinical Medical College of Fujian Medical University, Department of Urology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No.134, Dong Street, Fuzhou, 350001, People's Republic of China
| | - Deng Lin
- Shengli Clinical Medical College of Fujian Medical University, Department of Urology, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No.134, Dong Street, Fuzhou, 350001, People's Republic of China
| | - Yangpeng Lian
- Center for Information Management , Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No.134, Dong Street, Fuzhou, 350001, People's Republic of China
| | - Qichen Wei
- Department of Urology, Gutian County Hospital, Ningde, 352200, People's Republic of China
| | - Longbao Zheng
- Department of Urology, The 907nd Hospital of PLA, Nanping, 353000, Fujian, People's Republic of China
| | - Kun Yuan
- Department of Urology, Zhuzhou People's Hospital , Zhuzhou, People's Republic of China
| | - Jiayang Zhao
- Newland Education, Fuzhou, 350015, People's Republic of China
| | - Kaijin Kuang
- College of Finance, Fujian Jiang Xia University, Fuzhou, 350108, Fujian, People's Republic of China
| | - Yuanyuan Tang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Yunliang Gao
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Mental Disorders, Changsha, People's Republic of China.
- Hunan Clinical Research Center of Minimally Invasive Urology, Changsha, People's Republic of China.
| |
Collapse
|
14
|
Silva A, Mouchiroud M, Lavoie O, Beji S, Elmquist JK, Caron A. Liver adrenoceptor alpha-1b plays a key role in energy and glucose homeostasis in female mice. Am J Physiol Endocrinol Metab 2024; 327:E626-E635. [PMID: 39259165 PMCID: PMC11559639 DOI: 10.1152/ajpendo.00153.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024]
Abstract
The liver plays a major role in glucose and lipid homeostasis and acts as a key organ in the pathophysiology of metabolic diseases. Intriguingly, increased sympathetic nervous system (SNS) activity to the liver has been associated with the development and progression of type 2 diabetes and obesity. However, the precise mechanisms by which the SNS regulates hepatic metabolism remain to be defined. Although liver α1-adrenoceptors were suggested to play a role in glucose homeostasis, the specific subtypes involved are unknown mainly because of the limitations of pharmacological tools. Here, we generated and validated a novel mouse model allowing tissue-specific deletion of α-1b adrenoceptor (Adra1b) in hepatocytes to investigate the role of liver ADRA1B in energy and glucose metabolism. We found that selective deletion of Adra1b in mouse liver has limited metabolic impact in lean mice. However, loss of Adra1b in hepatocytes exacerbated diet-induced obesity, insulin resistance, and glucose intolerance in female, but not in male mice. In obese females, this was accompanied by reduced hepatic gluconeogenic capacity and reprogramming of gonadal adipose tissue with hyperleptinemia. Our data highlight sex-dependent mechanisms by which the SNS regulates energy and glucose homeostasis through liver ADRA1B.NEW & NOTEWORTHY The sympathetic nervous system plays an important role in regulating hepatic physiology and metabolism. However, the identity of the adrenoceptors involved in these effects is still elusive. Using CRISPR-Cas9, we developed a novel transgenic tool to study the role of liver α-1b adrenoceptor (ADRA1B). We show that ADRA1B plays a key role in mediating the effects of the sympathetic nervous system on hepatic metabolism, particularly in female mice.
Collapse
Affiliation(s)
- Anisia Silva
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada
- Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| | | | - Olivier Lavoie
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada
- Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| | - Sarra Beji
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada
- Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| | - Joel K Elmquist
- Department of Internal Medicine, Center for Hypothalamic Research , University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada
- Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| |
Collapse
|
15
|
Mravec B, Szantova M. The role of the nervous system in liver diseases. Hepatol Res 2024; 54:970-980. [PMID: 39392763 DOI: 10.1111/hepr.14125] [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: 08/07/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
The nervous system significantly participates in maintaining homeostasis, and modulating repair and regeneration processes in the liver. Moreover, the nervous system also plays an important role in the processes associated with the development and progression of liver disease, and can either potentiate or inhibit these processes. The aim of this review is to describe the mechanisms and pathways through which the nervous system influences the development and progression of liver diseases, such as alcohol-associated liver disease, nonalcoholic fatty liver disease, cholestatic liver disease, hepatitis, cirrhosis, and hepatocellular carcinoma. Possible therapeutic implications based on modulation of signals transduction between the nervous system and the liver are also discussed.
Collapse
Affiliation(s)
- Boris Mravec
- Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
- Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Maria Szantova
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
16
|
Fei B, Zhao Y, Wang J, Wen P, Li J, Tanaka M, Wang Z, Li S. Leveraging adrenergic receptor blockade for enhanced nonalcoholic fatty liver disease treatment via a biomimetic nanoplatform. J Nanobiotechnology 2024; 22:591. [PMID: 39342261 PMCID: PMC11438098 DOI: 10.1186/s12951-024-02864-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation, steatosis and fibrosis. Sympathetic nerves play a critical role in maintaining hepatic lipid homeostasis and regulating fibrotic progression through adrenergic receptors expressed by hepatocytes and hepatic stellate cells; however, the use of sympathetic nerve-focused strategies for the treatment of NAFLD is still in the infancy. Herein, a biomimetic nanoplatform with ROS-responsive and ROS-scavenging properties was developed for the codelivery of retinoic acid (RA) and the adrenoceptor antagonist labetalol (LA). The nanoplatform exhibited improved accumulation and sufficient drug release in the fibrotic liver, thereby achieving precise codelivery of drugs. Integration of adrenergic blockade effectively interrupted the vicious cycle of sympathetic nerves with hepatic stellate cells (HSCs) and hepatocytes, which not only combined with RA to restore HSCs to a quiescent state but also helped to reduce hepatic lipid accumulation. We demonstrated the excellent ability of the biomimetic nanoplatform to ameliorate liver inflammation, fibrosis and steatosis. Our work highlights the tremendous potential of a sympathetic nerve-focused strategy for the management of NAFLD and provides a promising nanoplatform for the treatment of NAFLD.
Collapse
Affiliation(s)
- Bingyuan Fei
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Jine Wang
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Panyue Wen
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Junjie Li
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Masaru Tanaka
- Institute for Materials Chemistry and Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China.
| | - Shuo Li
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China.
| |
Collapse
|
17
|
Wilasrusmee KT, Sitticharoon C, Keadkraichaiwat I, Maikaew P, Pongwattanapakin K, Chatree S, Sririwichitchai R, Churintaraphan M. Epigallocatechin gallate enhances sympathetic heart rate variability and decreases blood pressure in obese subjects: a randomized control trial. Sci Rep 2024; 14:21628. [PMID: 39285220 PMCID: PMC11405511 DOI: 10.1038/s41598-024-72269-3] [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: 03/11/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
This study aimed to investigate effects of epigallocatechin gallate (EGCG) on blood pressure (BP) and autonomic nervous system, indicated by 5-min heart rate variability (HRV) measurement in obese subjects, and determine correlations of BP with metabolic factors. In a double-blind, randomized controlled trial, obese subjects (n = 30) were randomly allocated to receive 150 mg EGCG (n = 15) or placebo (n = 15) twice a day without dietary restrictions. After 8-week EGCG treatment, systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP) significantly decreased, while the low-frequency (LF) to high-frequency power (HF) ratio (LF/HF ratio) significantly increased (P < 0.05 all), indicating a shift toward sympathetic dominance, either directly or indirectly after BP lowering. SBP had positive correlations with obesity parameters, leptin, insulin, and insulin resistance but had a negative correlation with insulin sensitivity. DBP was positively correlated with age and HF in normalized unit, but negatively correlated with height and LF in ms2. High-density lipoprotein cholesterol (HDL-C) was negatively correlated with SBP, DBP, and MAP reflecting its protective effect against elevated BP. In conclusion, the 8-week EGCG treatment decreased BP and increased the LF/HF ratio, reflecting increased sympathetic activity, either a direct EGCG effect or an indirect compensatory response following BP reduction.
Collapse
Affiliation(s)
- Kittikorn Tommy Wilasrusmee
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Chantacha Sitticharoon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Issarawan Keadkraichaiwat
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Pailin Maikaew
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Kitchaya Pongwattanapakin
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Saimai Chatree
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Rungnapa Sririwichitchai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| | - Malika Churintaraphan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok, 10700, Thailand
| |
Collapse
|
18
|
Lei Q, Jiang Z, Shao Y, Liu X, Li X. Stellate ganglion, inflammation, and arrhythmias: a new perspective on neuroimmune regulation. Front Cardiovasc Med 2024; 11:1453127. [PMID: 39328238 PMCID: PMC11424448 DOI: 10.3389/fcvm.2024.1453127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Current research on the stellate ganglion (SG) has shifted from merely understanding its role as a collection of neurons to recognizing its importance in immune regulation. As part of the autonomic nervous system (ANS), the SG plays a crucial role in regulating cardiovascular function, particularly cardiac sympathetic nerve activity. Abnormal SG function can lead to disordered cardiac electrical activity, which in turn affects heart rhythm stability. Studies have shown that excessive activity of the SG is closely related to the occurrence of arrhythmias, especially in the context of inflammation. Abnormal activity of the SG may trigger excessive excitation of the sympathetic nervous system (SNS) through neuroimmune mechanisms, thereby increasing the risk of arrhythmias. Simultaneously, the inflammatory response of the SG further aggravates this process, forming a vicious cycle. However, the causal relationship between SG, inflammation, and arrhythmias has not yet been fully clarified. Therefore, this article deeply explores the key role of the SG in arrhythmias and its complex relationship with inflammation, providing relevant clinical evidence. It indicates that interventions targeting SG function and inflammatory responses have potential in preventing and treating inflammation-related arrhythmias, offering a new perspective for cardiovascular disease treatment strategies.
Collapse
Affiliation(s)
- Qiulian Lei
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zefei Jiang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yu Shao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xinghong Liu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaoping Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Wang B, Hu Z, Cui L, Zhao M, Su Z, Jiang Y, Liu J, Zhao Y, Hou Y, Yang X, Zhang C, Guo B, Li D, Zhao L, Zheng S, Zhao Y, Yang W, Wang D, Yu S, Zhu S, Yan Y, Yuan G, Li K, Zhang W, Qin L, Zhang W, Sun F, Luo J, Zheng R. βAR-mTOR-lipin1 pathway mediates PKA-RIIβ deficiency-induced adipose browning. Theranostics 2024; 14:5316-5335. [PMID: 39267778 PMCID: PMC11388065 DOI: 10.7150/thno.97046] [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: 04/07/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Background: Enhancing white adipose tissue (WAT) browning combats obesity. The RIIβ subunit of cAMP-dependent protein kinase (PKA) is primarily expressed in the brain and adipose tissue. Deletion of the hypothalamic RIIβ gene centrally induces WAT browning, yet the peripheral mechanisms mediating this process remain unexplored. Methods: This study investigates the mechanisms underlying WAT browning in RIIβ-KO mice. Genetic approaches such as β3-adrenergic receptors (β3ARs) deletion and sympathetic denervation of WAT were utilized. Genome-wide transcriptomic sequencing and bioinformatic analysis were employed to identify potential mediators of WAT browning. siRNA assays were employed to knock down mTOR and lipin1 in vitro, while AAV-shRNAs were used for the same purpose in vivo. Results: We found that WAT browning substantially contributes to the lean and obesity-resistant phenotypes of RIIβ-KO mice. The WAT browning can be dampened by β3ARs deletion or WAT sympathetic denervation. We identified that adipocytic mTOR and lipin1 may act as mediators of the WAT browning. Inhibition of mTOR or lipin1 abrogates WAT browning and hinders the lean phenotype of RIIβ-KO mice. In human subcutaneous white adipocytes and mouse white adipocytes, β3AR stimulation can activate mTOR and causes lipin1 nuclear translocation; knockdown of mTOR and Lipin1 mitigates WAT browning-associated gene expression, impedes mitochondrial activity. Moreover, mTOR knockdown reduces lipin1 level and nuclear translocation, indicating that lipin1 may act downstream of mTOR. Additionally, in vivo knockdown of mTOR and Lipin1 diminished WAT browning and increased adiposity. Conclusions: The β3AR-activated mTOR-lipin1 axis mediates WAT browning, offering new insights into the molecular basis of PKA-regulated WAT browning. These findings provide potential adipose target candidates for the development of drugs to treat obesity.
Collapse
Affiliation(s)
- Bingwei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, China
| | - Zhiping Hu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Peking University, Beijing, China
- Present address: Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Long Cui
- Department of General Surgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Miao Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhijie Su
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yong Jiang
- Department of General Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yun Zhao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoning Yang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Chenyu Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Daotong Li
- National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, College of Food Science and Nutritional Engineering, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Shengmin Zheng
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Peking University, Beijing, China
| | - Yiguo Zhao
- Department of Gastrointestinal Surgery, Peking University International Hospital, Peking University, Beijing, China
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Siwang Yu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, Peking University, Beijing, China
| | - Shigong Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University, Beijing, China
| | - Geheng Yuan
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wenqiang Zhang
- College of Engineering, China Agricultural University, Beijing, China
| | - Lihua Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Weiguang Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
- Beijing Life Science Academy, Beijing, China
| |
Collapse
|
20
|
Costa-E-Sousa RH, Brooks VL. The growing complexity of the control of the hypothalamic pituitary thyroid axis and brown adipose tissue by leptin. VITAMINS AND HORMONES 2024; 127:305-362. [PMID: 39864945 DOI: 10.1016/bs.vh.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The balance between food intake and energy expenditure is precisely regulated to maintain adipose stores. Leptin, which is produced in and released from adipose in direct proportion to its size, is a major contributor to this control and initiates its homeostatic responses largely via binding to leptin receptors (LepR) in the hypothalamus. Decreases in hypothalamic LepR binding signals starvation, leading to hunger and reduced energy expenditure, whereas increases in hypothalamic LepR binding can suppress food intake and increase energy expenditure. However, large gaps persist in the specific hypothalamic sites and detailed mechanisms by which leptin increases energy expenditure, via the parallel activation of the hypothalamic pituitary thyroid (HPT) axis and brown adipose tissue (BAT). The purpose of this review is to develop a framework for the complex mechanisms and neurocircuitry. The core circuitry begins with leptin binding to receptors in the arcuate nucleus, which then sends projections to the paraventricular nucleus (to regulate the HPT axis) and the dorsomedial hypothalamus (to regulate BAT). We build on this core by layering complexities, including the intricate and unsettled regulation of arcuate proopiomelanocortin neurons by leptin and the changes that occur as the regulation of the HPT axis and BAT is engaged or modified by challenges such as starvation, hypothermia, obesity, and pregnancy.
Collapse
Affiliation(s)
- Ricardo H Costa-E-Sousa
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Virginia L Brooks
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
21
|
Zhou B, Chen Q, Zhang Q, Tian W, Chen T, Liu Z. Therapeutic potential of adipose-derived stem cell extracellular vesicles: from inflammation regulation to tissue repair. Stem Cell Res Ther 2024; 15:249. [PMID: 39113098 PMCID: PMC11304935 DOI: 10.1186/s13287-024-03863-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Inflammation is a key pathological feature of many diseases, disrupting normal tissue structure and resulting in irreversible damage. Despite the need for effective inflammation control, current treatments, including stem cell therapies, remain insufficient. Recently, extracellular vesicles secreted by adipose-derived stem cells (ADSC-EVs) have garnered attention for their significant anti-inflammatory properties. As carriers of bioactive substances, these vesicles have demonstrated potent capabilities in modulating inflammation and promoting tissue repair in conditions such as rheumatoid arthritis, osteoarthritis, diabetes, cardiovascular diseases, stroke, and wound healing. Consequently, ADSC-EVs are emerging as promising alternatives to conventional ADSC-based therapies, offering advantages such as reduced risk of immune rejection, enhanced stability, and ease of storage and handling. However, the specific mechanisms by which ADSC-EVs regulate inflammation under pathological conditions are not fully understood. This review discusses the role of ADSC-EVs in inflammation control, their impact on disease prognosis, and their potential to promote tissue repair. Additionally, it provides insights into future clinical research focused on ADSC-EV therapies for inflammatory diseases, which overcome some limitations associated with cell-based therapies.
Collapse
Affiliation(s)
- Bohuai Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiuyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Qiuwen Zhang
- The Affiliated Stomatological Hospital Southwest Medical University, Luzhou, 646000, China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tian Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Zhi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
Morgan HJN, Delfino HBP, Schavinski AZ, Malone SA, Charoy C, Reis NG, Assis AP, Lautherbach N, Silveira WA, Heck LC, Guton D, Domingos AI, Kettelhut IC, Montminy M, Navegantes LCC. Hepatic noradrenergic innervation acts via CREB/CRTC2 to activate gluconeogenesis during cold. Metabolism 2024; 157:155940. [PMID: 38878857 DOI: 10.1016/j.metabol.2024.155940] [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: 03/28/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND AND AIM Although it is well established that hormones like glucagon stimulates gluconeogenesis via the PKA-mediated phosphorylation of CREB and dephosphorylation of the cAMP-regulated CREB coactivators CRTC2, the role of neural signals in the regulation of gluconeogenesis remains uncertain. METHODS AND RESULTS Here, we characterize the noradrenergic bundle architecture in mouse liver; we show that the sympathoexcitation induced by acute cold exposure promotes hyperglycemia and upregulation of gluconeogenesis via triggering of the CREB/CRTC2 pathway. Following its induction by dephosphorylation, CRTC2 translocates to the nucleus and drives the transcription of key gluconeogenic genes. Rodents submitted to different models of sympathectomy or knockout of CRTC2 do not activate gluconeogenesis in response to cold. Norepinephrine directly acts in hepatocytes mainly through a Ca2+-dependent pathway that stimulates CREB/CRTC2, leading to activation of the gluconeogenic program. CONCLUSION Our data demonstrate the importance of the CREB/CRTC2 pathway in mediating effects of hepatic sympathetic inputs on glucose homeostasis, providing new insights into the role of norepinephrine in health and disease.
Collapse
Affiliation(s)
- Henrique J N Morgan
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Heitor B P Delfino
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Aline Z Schavinski
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Samuel A Malone
- Department of Physiology, Genetics and Anatomy, University of Oxford, Oxford, UK
| | | | - Natany G Reis
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana P Assis
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Natalia Lautherbach
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Wilian A Silveira
- Department of Biochemistry, Pharmacology and Physiology, Institute of Biological and Natural Sciences, Federal University of Triangulo Mineiro (UFTM), Uberaba, Minas Gerais, Brazil
| | - Lilian C Heck
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dan Guton
- The Francis Crick Institute, London, UK
| | - Ana I Domingos
- Department of Physiology, Genetics and Anatomy, University of Oxford, Oxford, UK
| | - Isis C Kettelhut
- Department of Biochemistry/Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marc Montminy
- Peptide Biology Laboratories, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Luiz C C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
23
|
Braga GDC, Simões JLB, Teixeira Dos Santos YJ, Filho JCM, Bagatini MD. The impacts of obesity in rheumatoid arthritis and insights into therapeutic purinergic modulation. Int Immunopharmacol 2024; 136:112357. [PMID: 38810303 DOI: 10.1016/j.intimp.2024.112357] [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: 04/22/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Rheumatoid Arthritis (RA) is an autoimmune condition responsible for the impairment of synovia and joints, endangering the functionality of individuals and contributing to mortality. Currently, obesity is increasing worldwide, and recent studies have suggested an association between such condition and RA. In this sense, obese individuals present a lower capacity for achieving remission and present more intense symptoms of the disease, demonstrating a link between both disorders. Different studies aim to understand the possible connection between the conditions; however, few is known in this sense. Therefore, knowing that obesity can alter the activity of multiple body systems, this work's objective is to evaluate the main modifications caused by obesity, which can be linked to the pathophysiology of RA, highlighting as relevant topics obesity's negative impact triggering systemic inflammation, intestinal dysbiosis, endocrine disbalances. Furthermore, the relationship between oxidative stress and obesity also deserves to be highlighted, considering the influence of reactive oxygen species (ROS) accumulation in RA exacerbation. Additionally, many of those characteristics influenced by obesity, along with the classic peculiarities of RA pathophysiology, can also be associated with purinergic signaling. Hence, this work suggests possible connections between the purinergic system and RA, proposing potential therapeutic targets against RA to be studied.
Collapse
|
24
|
Huang J, Liu X, Qiu Q, Tan W, Li R, Xi H, Peng C, Zhou L, Zhou X, Wang Y, Jiang H. Blockade of mesenteric and omental adipose tissue sensory neurons improves cardiac remodeling through sympathetic pathway. iScience 2024; 27:110245. [PMID: 39055939 PMCID: PMC11269788 DOI: 10.1016/j.isci.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 05/01/2024] [Accepted: 06/07/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenteric and omental adipose tissue (MOAT) communicates directly with the heart through the secretion of bioactive molecules and indirectly through afferent signaling to the central nervous system. Myocardial infarction (MI) may induce pathological alterations in MOAT, which further affects cardiac function. Our study revealed that MI induced significant MOAT transcriptional changes in genes related with signal transduction, including adiponectin (APN), neuropeptide Y (NPY), and complement C3 (C3), potentially influencing afferent activity. We further found that MOAT sensory nerve denervation with capsaicin (CAP) prevented cardiac remodeling, improved cardiac function, and reversed cardiac sympathetic nerve hyperactivation in the MI group, accompanied by reduced serum norepinephrine. In addition, CAP reversed the elevated MOAT afferent input and brain-heart sympathetic outflow post-MI, increasing APN and NPY and decreasing C3 and serum proinflammatory factors. These results demonstrated that blockade of the MOAT afferent sensory nerve exerts a cardioprotective effect by inhibiting the brain-heart sympathetic axis.
Collapse
Affiliation(s)
- Jiaxing Huang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Xinyu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Qinfang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Wuping Tan
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Rui Li
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Haosong Xi
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Chen Peng
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Yueyi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University; Hubei Key Laboratory of Autonomic Nervous System Modulation; Taikang Center for Life and Medical Sciences, Wuhan University; Cardiac Autonomic Nervous System Research Center of Wuhan University; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| |
Collapse
|
25
|
Cui Y, Auclair H, He R, Zhang Q. GPCR-mediated regulation of beige adipocyte formation: Implications for obesity and metabolic health. Gene 2024; 915:148421. [PMID: 38561165 DOI: 10.1016/j.gene.2024.148421] [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: 01/11/2024] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Obesity and its associated complications pose a significant burden on health. The non-shivering thermogenesis (NST) and metabolic capacity properties of brown adipose tissue (BAT), which are distinct from those of white adipose tissue (WAT), in combating obesity and its related metabolic diseases has been well documented. However, beige adipose tissue, the third and relatively novel type of adipose tissue, which emerges in extensive presence of WAT and shares similar favorable metabolic properties with BAT, has garnered considerable attention in recent years. In this review, we focused on the role of G protein-coupled receptors (GPCRs), the largest receptor family and the most successful class of drug targets in humans, in the induction of beige adipocytes. More importantly, we highlight researchers' clinical treatment attempts to ameliorate obesity and other related metabolic diseases through the formation and activation of beige adipose tissue. In summary, this review provides valuable insights into the formation of beige adipose tissue and the involvement of GPCRs, based on the latest advancements in scientific research.
Collapse
Affiliation(s)
- Yuanxu Cui
- Animal Zoology Department, Kunming Medical University, Kunming, China; Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Hugo Auclair
- Faculty of Medicine, François-Rabelais University, Tours, France
| | - Rong He
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Qiang Zhang
- Animal Zoology Department, Kunming Medical University, Kunming, China.
| |
Collapse
|
26
|
Zhang Z, Zhang D, Lin Q, Cui X. Therapeutically Fine-Tuning Autonomic Nervous System to Treat Sepsis: A New Perspective on the Immunomodulatory Effects of Acupuncture. J Inflamm Res 2024; 17:4373-4387. [PMID: 38988505 PMCID: PMC11233988 DOI: 10.2147/jir.s477181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Recent studies have highlighted the immunomodulatory effects of acupuncture on sepsis and proposed novel non-pharmacological or bioelectronic approaches to managing inflammatory illnesses. Establishing rules for selectively activating sympathetic or vagal nerve-mediated anti-inflammatory pathways using acupuncture has valuable clinical applications. Over the years, studies have revealed the segmental modulatory role of acupuncture in regulating visceral function by targeting the autonomic nervous system (ANS). In this review, we aim to summarize recent findings on acupuncture in treating sepsis, focusing on the underlying ANS mechanism, as well as the rules of acupoint specificity, intensity, frequency, and other parameters utilized in these studies. Mechanistically, the immunomodulatory properties of the sympathetic nervous system have been highlighted. Furthermore, we explore the immunotherapeutic benefits of acupuncture in treating sepsis. A better understanding of the immunoregulatory mechanism of sympathetic nervous system may offer novel approaches for the development of therapeutics to treat or prevent a variety of inflammatory diseases.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Dingdan Zhang
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, 21287, USA
| | - Xiang Cui
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, People’s Republic of China
| |
Collapse
|
27
|
Mandal SK, Yadav P, Sheth RA. The Neuroimmune Axis and Its Therapeutic Potential for Primary Liver Cancer. Int J Mol Sci 2024; 25:6237. [PMID: 38892423 PMCID: PMC11172507 DOI: 10.3390/ijms25116237] [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: 02/05/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The autonomic nervous system plays an integral role in motion and sensation as well as the physiologic function of visceral organs. The nervous system additionally plays a key role in primary liver diseases. Until recently, however, the impact of nerves on cancer development, progression, and metastasis has been unappreciated. This review highlights recent advances in understanding neuroanatomical networks within solid organs and their mechanistic influence on organ function, specifically in the liver and liver cancer. We discuss the interaction between the autonomic nervous system, including sympathetic and parasympathetic nerves, and the liver. We also examine how sympathetic innervation affects metabolic functions and diseases like nonalcoholic fatty liver disease (NAFLD). We also delve into the neurobiology of the liver, the interplay between cancer and nerves, and the neural regulation of the immune response. We emphasize the influence of the neuroimmune axis in cancer progression and the potential of targeted interventions like neurolysis to improve cancer treatment outcomes, especially for hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
| | | | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1471, Houston, TX 77030-4009, USA; (S.K.M.); (P.Y.)
| |
Collapse
|
28
|
Rahmouni K. Neural Circuits Underlying Reciprocal Cardiometabolic Crosstalk: 2023 Arthur C. Corcoran Memorial Lecture. Hypertension 2024; 81:1233-1243. [PMID: 38533662 PMCID: PMC11096079 DOI: 10.1161/hypertensionaha.124.22066] [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] [Indexed: 03/28/2024]
Abstract
The interplay of various body systems, encompassing those that govern cardiovascular and metabolic functions, has evolved alongside the development of multicellular organisms. This evolutionary process is essential for the coordination and maintenance of homeostasis and overall health by facilitating the adaptation of the organism to internal and external cues. Disruption of these complex interactions contributes to the development and progression of pathologies that involve multiple organs. Obesity-associated cardiovascular risks, such as hypertension, highlight the significant influence that metabolic processes exert on the cardiovascular system. This cardiometabolic communication is reciprocal, as indicated by substantial evidence pointing to the ability of the cardiovascular system to affect metabolic processes, with pathophysiological implications in disease conditions. In this review, I outline the bidirectional nature of the cardiometabolic interaction, with special emphasis on the impact that metabolic organs have on the cardiovascular system. I also discuss the contribution of the neural circuits and autonomic nervous system in mediating the crosstalk between cardiovascular and metabolic functions in health and disease, along with the molecular mechanisms involved.
Collapse
Affiliation(s)
- Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
29
|
Chen ZT, Weng ZX, Lin JD, Meng ZX. Myokines: metabolic regulation in obesity and type 2 diabetes. LIFE METABOLISM 2024; 3:loae006. [PMID: 39872377 PMCID: PMC11749576 DOI: 10.1093/lifemeta/loae006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 01/30/2025]
Abstract
Skeletal muscle plays a vital role in the regulation of systemic metabolism, partly through its secretion of endocrine factors which are collectively known as myokines. Altered myokine levels are associated with metabolic diseases, such as type 2 diabetes (T2D). The significance of interorgan crosstalk, particularly through myokines, has emerged as a fundamental aspect of nutrient and energy homeostasis. However, a comprehensive understanding of myokine biology in the setting of obesity and T2D remains a major challenge. In this review, we discuss the regulation and biological functions of key myokines that have been extensively studied during the past two decades, namely interleukin 6 (IL-6), irisin, myostatin (MSTN), growth differentiation factor 11 (GDF11), fibroblast growth factor 21 (FGF21), apelin, brain-derived neurotrophic factor (BDNF), meteorin-like (Metrnl), secreted protein acidic and rich in cysteine (SPARC), β-aminoisobutyric acid (BAIBA), Musclin, and Dickkopf 3 (Dkk3). Related to these, we detail the role of exercise in myokine expression and secretion together with their contributions to metabolic physiology and disease. Despite significant advancements in myokine research, many myokines remain challenging to measure accurately and investigate thoroughly. Hence, new research techniques and detection methods should be developed and rigorously tested. Therefore, developing a comprehensive perspective on myokine biology is crucial, as this will likely offer new insights into the pathophysiological mechanisms underlying obesity and T2D and may reveal novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zhi-Tian Chen
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute (ZJE), School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Zhi-Xuan Weng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
30
|
Wang Y, Ye L. The Afferent Function of Adipose Innervation. Diabetes 2024; 73:348-354. [PMID: 38377447 PMCID: PMC10882147 DOI: 10.2337/dbi23-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/18/2023] [Indexed: 02/22/2024]
Abstract
Adipose tissue innervation is critical for regulating metabolic and energy homeostasis. While the sympathetic efferent innervation of fat is well characterized, the role of sensory or afferent innervation remains less explored. This article reviews previous work on adipose innervation and recent advances in the study of sensory innervation of adipose tissues. We discuss key open questions, including the physiological implications of adipose afferents in homeostasis as well as potential cross talk with sympathetic neurons, the immune system, and hormonal pathways. We also outline the general technical challenges of studying dorsal root ganglia innervating fat, along with emerging technologies that may overcome these barriers. Finally, we highlight areas for further research to deepen our understanding of the afferent function of adipose innervation.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
31
|
Ren W, Hua M, Cao F, Zeng W. The Sympathetic-Immune Milieu in Metabolic Health and Diseases: Insights from Pancreas, Liver, Intestine, and Adipose Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306128. [PMID: 38039489 PMCID: PMC10885671 DOI: 10.1002/advs.202306128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/28/2023] [Indexed: 12/03/2023]
Abstract
Sympathetic innervation plays a crucial role in maintaining energy balance and contributes to metabolic pathophysiology. Recent evidence has begun to uncover the innervation landscape of sympathetic projections and sheds light on their important functions in metabolic activities. Additionally, the immune system has long been studied for its essential roles in metabolic health and diseases. In this review, the aim is to provide an overview of the current research progress on the sympathetic regulation of key metabolic organs, including the pancreas, liver, intestine, and adipose tissues. In particular, efforts are made to highlight the critical roles of the peripheral nervous system and its potential interplay with immune components. Overall, it is hoped to underscore the importance of studying metabolic organs from a comprehensive and interconnected perspective, which will provide valuable insights into the complex mechanisms underlying metabolic regulation and may lead to novel therapeutic strategies for metabolic diseases.
Collapse
Affiliation(s)
- Wenran Ren
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Meng Hua
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Fang Cao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhou563000China
| | - Wenwen Zeng
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineTaiyuan030001China
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijing100084China
| |
Collapse
|
32
|
Haberman ER, Sarker G, Arús BA, Ziegler KA, Meunier S, Martínez-Sánchez N, Freibergerová E, Yilmaz-Özcan S, Fernández-González I, Zentai C, O'Brien CJO, Grainger DE, Sidarta-Oliveira D, Chakarov S, Raimondi A, Iannacone M, Engelhardt S, López M, Ginhoux F, Domingos AI. Immunomodulatory leptin receptor + sympathetic perineurial barrier cells protect against obesity by facilitating brown adipose tissue thermogenesis. Immunity 2024; 57:141-152.e5. [PMID: 38091996 DOI: 10.1016/j.immuni.2023.11.006] [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: 05/31/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/12/2024]
Abstract
Adipose tissues (ATs) are innervated by sympathetic nerves, which drive reduction of fat mass via lipolysis and thermogenesis. Here, we report a population of immunomodulatory leptin receptor-positive (LepR+) sympathetic perineurial barrier cells (SPCs) present in mice and humans, which uniquely co-express Lepr and interleukin-33 (Il33) and ensheath AT sympathetic axon bundles. Brown ATs (BATs) of mice lacking IL-33 in SPCs (SPCΔIl33) had fewer regulatory T (Treg) cells and eosinophils, resulting in increased BAT inflammation. SPCΔIl33 mice were more susceptible to diet-induced obesity, independently of food intake. Furthermore, SPCΔIl33 mice had impaired adaptive thermogenesis and were unresponsive to leptin-induced rescue of metabolic adaptation. We therefore identify LepR+ SPCs as a source of IL-33, which orchestrate an anti-inflammatory BAT environment, preserving sympathetic-mediated thermogenesis and body weight homeostasis. LepR+IL-33+ SPCs provide a cellular link between leptin and immune regulation of body weight, unifying neuroendocrinology and immunometabolism as previously disconnected fields of obesity research.
Collapse
Affiliation(s)
- Emma R Haberman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Gitalee Sarker
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bernardo A Arús
- Instituto Gulbenkian de Ciência, Oeiras, Portugal; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Karin A Ziegler
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sandro Meunier
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Noelia Martínez-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Eliška Freibergerová
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Iara Fernández-González
- Neurobesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
| | - Chloe Zentai
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Conan J O O'Brien
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David E Grainger
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - Svetoslav Chakarov
- Singapore Immunology Network (SIgN), A(∗)STAR, Singapore, Singapore; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | - Matteo Iannacone
- Vita-Salute San Raffaele University, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University Munich (TUM), Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Miguel López
- Neurobesity Group, Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, Santiago, Spain
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A(∗)STAR, Singapore, Singapore; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
33
|
Muntjewerff EM, Josyula VS, Christoffersson G. Three-dimensional Co-culture Model for Live Imaging of Pancreatic Islets, Immune Cells, and Neurons in Agarose Gel. Bio Protoc 2023; 13:e4852. [PMID: 37900103 PMCID: PMC10603256 DOI: 10.21769/bioprotoc.4852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 10/31/2023] Open
Abstract
During the onset of autoimmune diabetes, nerve-immune cell interactions seem to play an important role; however, there are currently no models to follow and interfere with these interactions over time in vivo or in vitro. Two-dimensional in vitro models provide insufficient information and microfluidics or organs on a chip are usually challenging to work with. We present here what we believe to be the first simple model that provides the opportunity to co-culture pancreatic islets with sympathetic nerves and immune cells. This model is based on our stamping device that can be 3D printed (STL file provided). Due to the imprint in the agarose gel, sympathetic neurons, pancreatic islets, and macrophages can be seeded in specific locations at a level that allows for confocal live-cell imaging. In this protocol, we provide the instructions to construct and perform live cell imaging experiments in our co-culture model, including: 1) design for the stamping device to make the imprint in the gel, 2) isolation of sympathetic neurons, pancreatic islets, and macrophages, 3) co-culture conditions, 4) how this can be used for live cell imaging, and 5) possibilities for wider use of the model. In summary, we developed an easy-to-use co-culture model that allows manipulation and imaging of interactions between sympathetic nerves, pancreatic islets, and macrophages. This new co-culture model is useful to study nerve-immune cell-islet interactions and will help to identify the functional relevance of neuro-immune interactions in the pancreas. Key features • A novel device that allows for 3D co-culture of sympathetic neurons, pancreatic islets, and immune cells • The device allows the capture of live interactions between mouse sympathetic neurons, pancreatic islets, and immune cells in a controlled environment after six days of co-culturing. • This protocol uses cultured sympathetic neurons isolated from the superior cervical ganglia using a previously established method (Jackson and Tourtellotte, 2014) in a 3D co-culture. • This method requires 3D printing of our own designed gel-stamping device (STL print file provided on SciLifeLab FigShare DOI: 10.17044/scilifelab.24073062).
Collapse
Affiliation(s)
| | - Vijay S Josyula
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Gustaf Christoffersson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
34
|
Stabile AM, Pistilli A, Mariangela R, Rende M, Bartolini D, Di Sante G. New Challenges for Anatomists in the Era of Omics. Diagnostics (Basel) 2023; 13:2963. [PMID: 37761332 PMCID: PMC10529314 DOI: 10.3390/diagnostics13182963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023] Open
Abstract
Anatomic studies have traditionally relied on macroscopic, microscopic, and histological techniques to investigate the structure of tissues and organs. Anatomic studies are essential in many fields, including medicine, biology, and veterinary science. Advances in technology, such as imaging techniques and molecular biology, continue to provide new insights into the anatomy of living organisms. Therefore, anatomy remains an active and important area in the scientific field. The consolidation in recent years of some omics technologies such as genomics, transcriptomics, proteomics, and metabolomics allows for a more complete and detailed understanding of the structure and function of cells, tissues, and organs. These have been joined more recently by "omics" such as radiomics, pathomics, and connectomics, supported by computer-assisted technologies such as neural networks, 3D bioprinting, and artificial intelligence. All these new tools, although some are still in the early stages of development, have the potential to strongly contribute to the macroscopic and microscopic characterization in medicine. For anatomists, it is time to hitch a ride and get on board omics technologies to sail to new frontiers and to explore novel scenarios in anatomy.
Collapse
Affiliation(s)
- Anna Maria Stabile
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
| | - Alessandra Pistilli
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
| | - Ruggirello Mariangela
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
| | - Desirée Bartolini
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Gabriele Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 60132 Perugia, Italy; (A.M.S.); (A.P.); (R.M.); (M.R.)
| |
Collapse
|
35
|
Lund J, Clemmensen C. Physiological protection against weight gain: evidence from overfeeding studies and future directions. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220229. [PMID: 37482786 PMCID: PMC10363696 DOI: 10.1098/rstb.2022.0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/24/2023] [Indexed: 07/25/2023] Open
Abstract
Body weight is under physiological regulation. When body fat mass decreases, a series of responses are triggered to promote weight regain by increasing food intake and decreasing energy expenditure. Analogous, in response to experimental overfeeding, excessive weight gain is counteracted by a reduction in food intake and possibly by an increase in energy expenditure. While low blood leptin and other hormones defend against weight loss, the signals that oppose overfeeding-induced fat mass expansion are still unknown. In this article, we discuss insights gained from overfeeding interventions in humans and intragastric overfeeding studies in rodents. We summarize the knowledge on the relative contributions of energy intake, energy expenditure and energy excretion to the physiological defence against overfeeding-induced weight gain. Furthermore, we explore literature supporting the existence of unidentified endocrine and non-endocrine pathways that defend against weight gain. Finally, we discuss the physiological drivers of constitutional thinness and suggest that overfeeding of individuals with constitutional thinness represents a gateway to understand the physiology of weight gain resistance in humans. Experimental overfeeding, combined with modern multi-omics techniques, has the potential to unveil the long-sought signalling pathways that protect against weight gain. Discovering these mechanisms could give rise to new treatments for obesity. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
36
|
Fried SK. Adipose 'neighborhoods' collaborate to maintain metabolic health. Curr Opin Genet Dev 2023; 81:102079. [PMID: 37406429 PMCID: PMC10867982 DOI: 10.1016/j.gde.2023.102079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023]
Abstract
Body fat is stored in anatomically distinct adipose depots that vary in their cell composition and play specialized roles in systemic metabolic homeostasis via secreted products. Their local effects on nearby tissues (e.g. the gut and visceral adipose tissues) are increasingly recognized and this local crosstalk is being elucidated. The major subcutaneous fat depots, abdominal and gluteal-femoral, exert opposite effects on the risk of metabolic disease. The pace of research into developmental, sex, and genetic determinants of human adipose depot growth and function is rapidly accelerating, providing insight into the pathogenesis of metabolic dysfunction in persons with obesity.
Collapse
Affiliation(s)
- Susan K Fried
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1152, New York, NY 10029, USA.
| |
Collapse
|
37
|
Wang Y, Ye L. Somatosensory innervation of adipose tissues. Physiol Behav 2023; 265:114174. [PMID: 36965573 PMCID: PMC11537203 DOI: 10.1016/j.physbeh.2023.114174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
The increasing prevalence of obesity and type 2 diabetes has led to a greater interest in adipose tissue physiology. Adipose tissue is now understood as an organ with endocrine and thermogenic capacities in addition to its role in fat storage. It plays a critical role in systemic metabolism and energy regulation, and its activity is tightly regulated by the nervous system. Fat is now recognized to receive sympathetic innervation, which transmits information from the brain, as well as sensory innervation, which sends information into the brain. The role of sympathetic innervation in adipose tissue has been extensively studied. However, the extent and the functional significance of sensory innervation have long been unclear. Recent studies have started to reveal that sensory neurons robustly innervate adipose tissue and play an important role in regulating fat activity. This brief review will discuss both historical evidence and recent advances, as well as important remaining questions about the sensory innervation of adipose tissue.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
38
|
Li S, Yuan H, Yang K, Li Q, Xiang M. Pancreatic sympathetic innervation disturbance in type 1 diabetes. Clin Immunol 2023; 250:109319. [PMID: 37024024 DOI: 10.1016/j.clim.2023.109319] [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: 01/04/2023] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023]
Abstract
Pancreatic sympathetic innervation can directly affect the function of islet. The disorder of sympathetic innervation in islets during the occurrence of type 1 diabetes (T1D) has been reported to be controversial with the inducing factor unclarified. Several studies have uncovered the critical role that sympathetic signals play in controlling the local immune system. The survival and operation of endocrine cells can be regulated by immune cell infiltration in islets. In the current review, we focused on the impact of sympathetic signals working on islets cell regulation, and discussed the potential factors that can induce the sympathetic innervation disorder in the islets. We also summarized the effect of interference with the islet sympathetic signals on the T1D occurrence. Overall, complete understanding of the regulatory effect of sympathetic signals on islet cells and local immune system could facilitate to design better strategies to control inflammation and protect β cells in T1D therapy.
Collapse
Affiliation(s)
- Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huimin Yuan
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Keshan Yang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
39
|
Donnelly RP. Cytokines 2022: 10th Annual Meeting of the International Cytokine & Interferon Society. J Interferon Cytokine Res 2023; 43:55-58. [PMID: 36695718 DOI: 10.1089/jir.2022.29050.rad] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Raymond P Donnelly
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
40
|
Lavoie O, Michael NJ, Caron A. A critical update on the leptin-melanocortin system. J Neurochem 2023; 165:467-486. [PMID: 36648204 DOI: 10.1111/jnc.15765] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/25/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
The discovery of leptin in 1994 was an "eureka moment" in the field of neurometabolism that provided new opportunities to better understand the central control of energy balance and glucose metabolism. Rapidly, a prevalent model in the field emerged that pro-opiomelanocortin (POMC) neurons were key in promoting leptin's anorexigenic effects and that the arcuate nucleus of the hypothalamus (ARC) was a key region for the regulation of energy homeostasis. While this model inspired many important discoveries, a growing body of literature indicates that this model is now outdated. In this review, we re-evaluate the hypothalamic leptin-melanocortin model in light of recent advances that directly tackle previous assumptions, with a particular focus on the ARC. We discuss how segregated and heterogeneous these neurons are, and examine how the development of modern approaches allowing spatiotemporal, intersectional, and chemogenetic manipulations of melanocortin neurons has allowed a better definition of the complexity of the leptin-melanocortin system. We review the importance of leptin in regulating glucose homeostasis, but not food intake, through direct actions on ARC POMC neurons. We further highlight how non-POMC, GABAergic neurons mediate leptin's direct effects on energy balance and influence POMC neurons.
Collapse
Affiliation(s)
- Olivier Lavoie
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada.,Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| | - Natalie Jane Michael
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada.,Quebec Heart and Lung Institute, Quebec City, Quebec, Canada
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, Quebec, Canada.,Quebec Heart and Lung Institute, Quebec City, Quebec, Canada.,Montreal Diabetes Research Center, Montreal, Quebec, Canada
| |
Collapse
|
41
|
Gagnon M, Fournier S, Marcouiller F, Guay L, Joseph V, Michael NJ, Kinkead R. Intermittent Hypoxia and Weight Loss: Insights into the Etiology of the Sleep Apnea Phenotype. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:61-71. [PMID: 37322336 DOI: 10.1007/978-3-031-32371-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Sleep apnea (SA) is a major respiratory disorder with increased risk for hypertension and obesity; however, our understanding of the origins of this complex disorder remains limited. Because apneas lead to recurrent drops in O2 during sleep, intermittent hypoxia (IH) is the main animal model to explore the pathophysiology of SA. Here, we assessed the impacts of IH on metabolic function and related signals. Adult male rats were exposed to 1 week of moderate IH (FiO2 = 0.10-30 s, ten cycles/hour, 8 h/day). Using whole-body plethysmography, we measured respiratory variability and apnea index during sleep. Blood pressure and heart rate were measured by the tail-cuff method; blood samples were taken for multiplex assay. At rest, IH augmented arterial blood pressure, respiratory instability, but not apnea index. IH induced weight, fat, and fluid loss. IH also reduced food intake and plasma leptin, adrenocorticotropic hormone (ACTH), and testosterone levels but increased inflammatory cytokines. We conclude that IH does not replicate the metabolic clinical features of SA patient, thus raising our awareness of the limitations of the IH model. The fact that the risk for hypertension occurs before the appearance of apneas provides new insights into the progression of the disease.
Collapse
Affiliation(s)
- Marianne Gagnon
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada.
- Faculty of Pharmacy, Université Laval, Québec City, QC, Canada.
| | - Stéphanie Fournier
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - Loralie Guay
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Natalie J Michael
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Richard Kinkead
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
42
|
Xia S, Yu X, Chen G. Pain as a Protective Factor for Alzheimer Disease in Patients with Cancer. Cancers (Basel) 2022; 15:cancers15010248. [PMID: 36612244 PMCID: PMC9818585 DOI: 10.3390/cancers15010248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Alzheimer disease (AD) and cancer have been reported to be inversely correlated in incidence, but the mechanism remains elusive. METHODS A case-control study was conducted, based on the SEER (Surveillance, Epidemiology, and End Results) Research Plus data, to evaluate 12 factors in patients with cancer. RESULTS Severe pain was related to reduced AD risk, while older age at cancer diagnosis, female, longer survival years after tumor diagnosis, more benign/borderline tumors, less cancer-directed surgery, and more chemotherapy were associated with higher AD risk. In addition, patients of different races or with different cancer sites were associated with different risks of getting AD. Cases had a higher prevalence of severe pain than controls in all race and cancer site subgroups, except for in digestive cancer, where the result was the opposite. CONCLUSIONS This study indicated pain as a novel protective factor for AD in patients with cancer. The mechanism behind it may provide new perspective on AD pathogenesis and AD-cancer association, which we discussed in our own hypothesis of the mechanism of pain action. In addition, digestive cancer pain had an opposite impact on AD risk from other cancer pains, which suggests the uniqueness of digestive system in interacting with the central nervous system.
Collapse
Affiliation(s)
- Siqi Xia
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Zhejiang University, Hangzhou 310003, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310003, China
| | - Xiaobo Yu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Zhejiang University, Hangzhou 310003, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310003, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Zhejiang University, Hangzhou 310003, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou 310003, China
- Correspondence: ; Tel.: +86-1380-5716-226
| |
Collapse
|