1
|
Park CB, Lee CH, Kang GM, Min SH, Kim MS. Microglial progranulin differently regulates hypothalamic lysosomal function in lean and obese conditions via cleavage-dependent mechanisms. J Neuroinflammation 2025; 22:68. [PMID: 40055725 PMCID: PMC11887206 DOI: 10.1186/s12974-025-03370-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/06/2025] [Indexed: 05/13/2025] Open
Abstract
Progranulin (PGRN) is a secretory precursor protein composed of 7.5 granulins (GRNs). Mutations in the PGRN-encoding gene Grn have been associated with neurodegenerative diseases. In our previous study, we found that Grn depletion in microglia disrupted glucose metabolism in mice fed a normal chow diet (NCD) but prevented the development of obesity in mice on a high-fat diet (HFD). Given that PGRN regulates lysosomal functions, we investigated lysosomal changes in the hypothalamus of mice with microglia-specific Grn depletion. Here we report that microglia-specific Grn depletion affects the lysosomes of hypothalamic proopiomelanocortin (POMC) neurons and microglia in diet-dependent fashion. Under NCD conditions, microglial Grn depletion led to increased lysosome mass, reduced lysosomal degradative capacity, and accumulation of lipofuscin and cytoplasmic TDP-43 in hypothalamic cells, indicative of lysosomal stress and dysfunction. In contrast, under HFD conditions, the absence of microglial Grn suppressed HFD-induced hypothalamic lysosomal stress. In cultured hypothalamic neurons and microglia, PGRN treatment enhanced lysosomal function, an effect inhibited by PGRN cleavage but restored when its cleavage was blocked. Since HFD feeding promotes the cleavage of hypothalamic PGRN into multi-GRNs and GRNs, the diet-dependent lysosomal changes observed in microglial Grn-depleted mice may be linked to PGRN cleavage. We also demonstrated that intracerebroventricular injection of bafilomycin, which induces lysosomal stress, resulted in microglial activation, inflammation, disrupted POMC neuronal circuitry, and impaired leptin signaling in the hypothalamus-common features of obesity. Our results indicate that microglial PGRN plays an important role in maintaining hypothalamic lysosomal function under healthy diet conditions, whereas increased cleavage of microglial PGRN in states of overnutrition disrupts hypothalamic lysosomal function, thereby fostering hypothalamic inflammation and obesity.
Collapse
Affiliation(s)
- Chae Beom Park
- Department of Biomedical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Chan Hee Lee
- Department of Biomedical Sciences, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Gil Myoung Kang
- Asan Institute for Life Science, Asan Medical Center , Seoul, 05505, Republic of Korea
| | - Se Hee Min
- Diabetes Center, Asan Medical Center, Seoul, 05505, Republic of Korea
- Division of Endocrinology and Metabolism, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Min-Seon Kim
- Diabetes Center, Asan Medical Center, Seoul, 05505, Republic of Korea.
- Division of Endocrinology and Metabolism, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
2
|
Placzek M, Chinnaiya K, Kim DW, Blackshaw S. Control of tuberal hypothalamic development and its implications in metabolic disorders. Nat Rev Endocrinol 2025; 21:118-130. [PMID: 39313573 PMCID: PMC11864813 DOI: 10.1038/s41574-024-01036-1] [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] [Accepted: 08/29/2024] [Indexed: 09/25/2024]
Abstract
The tuberal hypothalamus regulates a range of crucial physiological processes, including energy homeostasis and metabolism. In this Review, we explore the intricate molecular mechanisms and signalling pathways that control the development of the tuberal hypothalamus, focusing on aspects that shape metabolic outcomes. Major developmental events are discussed in the context of their effect on the establishment of both functional hypothalamic neuronal circuits and brain-body interfaces that are pivotal to the control of metabolism. Emerging evidence indicates that aberrations in molecular pathways during tuberal hypothalamic development contribute to metabolic dysregulation. Understanding the molecular underpinnings of tuberal hypothalamic development provides a comprehensive view of neurodevelopmental processes and offers a promising avenue for future targeted interventions to prevent and treat metabolic disorders.
Collapse
Affiliation(s)
- Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK.
- Bateson Centre, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
| | | | - Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Melnyk O, Guo JK, Li ZA, Jo JH, Hughes JW, Linnemann AK. Intravital imaging reveals glucose-dependent cilia movement in pancreatic islets in vivo. Metabolism 2025; 163:156105. [PMID: 39667431 PMCID: PMC11718731 DOI: 10.1016/j.metabol.2024.156105] [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: 08/14/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Pancreatic islet cells harbor primary cilia, small sensory organelles that detect environmental changes to regulate hormone secretion and intercellular communication. While the sensory and signaling capacity of primary cilia are well-appreciated, it is less recognized that these organelles also possess active motility, including in dense multicellular tissues such as the pancreatic islet. In this manuscript, we use transgenic cilia reporter mice and an intravital imaging approach to quantitate primary cilia dynamics as it occurs in live mouse pancreatic islets. We validate this imaging workflow as suitable for studying islet cilia motion in real time in vivo and demonstrate that glucose stimulation corresponds to a change in cilia motility, which may be a physiologic measure of nutrient-dependent fluxes in islet cell function. Complementary ex vivo analysis of isolated islets further demonstrates that metabolic stress in the form of lipotoxicity impairs cilia motility and these effects can be reversed by glucose elevation. These findings suggest that cilia motility is sensitive to metabolic stress and highlight its potential functional role in beta cell adaptation.
Collapse
Affiliation(s)
- Olha Melnyk
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeff Kaihao Guo
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zipeng Alex Li
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeong Hun Jo
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Jing W Hughes
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, St. Louis, MO, USA.
| | - Amelia K Linnemann
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
4
|
Collet TH, Schwitzgebel V. Exploring the therapeutic potential of precision medicine in rare genetic obesity disorders: a scientific perspective. Front Nutr 2024; 11:1509994. [PMID: 39777073 PMCID: PMC11705004 DOI: 10.3389/fnut.2024.1509994] [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: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The prevalence of obesity is increasing worldwide, affecting both children and adults. This obesity epidemic is mostly driven by an increase in energy intake (abundance of highly palatable energy-dense food and drinks) and to a lesser degree a decrease in energy expenditure (sedentary lifestyle). A small proportion of individuals with obesity are affected by genetic forms of obesity, which often relate to mutations in the leptin-melanocortin pathway or are part of syndromes such as the Bardet-Biedl syndrome. These rare forms of obesity have provided valuable insights into the genetic architecture of obesity. Recent advances in understanding the molecular mechanisms that control appetite, hunger, and satiety have led to the development of drugs that can override genetic defects, enabling precision treatment. Leptin deficiency is uniquely treated with recombinant human metreleptin, while those with LEPR, PCSK1, or POMC deficiency can now be treated with the MC4R agonist setmelanotide. This review highlights the most frequent monogenic and syndromic forms of obesity, and the future outlook of precision treatment for these conditions.
Collapse
Affiliation(s)
- Tinh-Hai Collet
- Service of Endocrinology, Diabetes, Nutrition, and Therapeutic Education, Geneva University Hospitals, Geneva, Switzerland
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
| | - Valerie Schwitzgebel
- Faculty of Medicine, Diabetes Center, University of Geneva, Geneva, Switzerland
- Pediatric Endocrine and Diabetes Unit, Department of Pediatrics, Obstetrics, and Gynecology, Geneva University Hospitals, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| |
Collapse
|
5
|
García-Navarrete C, Kretschmar C, Toledo J, Gutiérrez K, Hernández-Cáceres MP, Budini M, Parra V, Burgos PV, Lavandero S, Morselli E, Peña-Oyarzún D, Criollo A. PKD2 regulates autophagy and forms a protein complex with BECN1 at the primary cilium of hypothalamic neuronal cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167256. [PMID: 38782303 DOI: 10.1016/j.bbadis.2024.167256] [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: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
The primary cilium, hereafter cilium, is an antenna-like organelle that modulates intracellular responses, including autophagy, a lysosomal degradation process essential for cell homeostasis. Dysfunction of the cilium is associated with impairment of autophagy and diseases known as "ciliopathies". The discovery of autophagy-related proteins at the base of the cilium suggests its potential role in coordinating autophagy initiation in response to physiopathological stimuli. One of these proteins, beclin-1 (BECN1), it which is necessary for autophagosome biogenesis. Additionally, polycystin-2 (PKD2), a calcium channel enriched at the cilium, is required and sufficient to induce autophagy in renal and cancer cells. We previously demonstrated that PKD2 and BECN1 form a protein complex at the endoplasmic reticulum in non-ciliated cells, where it initiates autophagy, but whether this protein complex is present at the cilium remains unknown. Anorexigenic pro-opiomelanocortin (POMC) neurons are ciliated cells that require autophagy to maintain intracellular homeostasis. POMC neurons are sensitive to metabolic changes, modulating signaling pathways crucial for controlling food intake. Exposure to the saturated fatty acid palmitic acid (PA) reduces ciliogenesis and inhibits autophagy in these cells. Here, we show that PKD2 and BECN1 form a protein complex in N43/5 cells, an in vitro model of POMC neurons, and that both PKD2 and BECN1 locate at the cilium. In addition, our data show that the cilium is required for PKD2-BECN1 protein complex formation and that PA disrupts the PKD2-BECN1 complex, suppressing autophagy. Our findings provide new insights into the mechanisms by which the cilium controls autophagy in hypothalamic neuronal cells.
Collapse
Affiliation(s)
- Camila García-Navarrete
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Catalina Kretschmar
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Jorge Toledo
- Advanced Scientific Equipment Network (REDECA), Facultad de Medicina, Universidad de Chile, Chile
| | - Karla Gutiérrez
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - María Paz Hernández-Cáceres
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile; Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Mauricio Budini
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile; Autophagy Research Center, Santiago, Chile
| | - Valentina Parra
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas & Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas & Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Patricia V Burgos
- Autophagy Research Center, Santiago, Chile; Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile; Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Avenida Del Valle Norte #725, Huechuraba 8580702, Santiago, Chile
| | - Sergio Lavandero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas & Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas & Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile; Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile; Autophagy Research Center, Santiago, Chile
| | - Daniel Peña-Oyarzún
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas & Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile; Faculty of Odontology and Rehabilitation Sciences, Universidad San Sebastián, Chile.
| | - Alfredo Criollo
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile; Autophagy Research Center, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas & Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
6
|
Gallo R, Teijeiro A, Angulo-Aguado M, Djouder N. IL-17A produced by POMC neurons regulates diet-induced obesity. iScience 2024; 27:110259. [PMID: 39027371 PMCID: PMC11255842 DOI: 10.1016/j.isci.2024.110259] [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: 04/27/2023] [Revised: 08/29/2023] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Overeating leads to obesity, a low-grade inflammatory condition involving interleukin-17A (IL-17A). While pro-opiomelanocortin (POMC) neurons regulate feeding, their connection with IL-17A is not well understood. To impair IL-17A signaling in POMC neurons, IL-17A receptor (Il17ra) was deleted by crossing IL17ra-flox and Pomc-Cre mice. Despite effective deletion, these mice showed no differences in body weight or adiposity compared to control mice, challenging the idea that IL-17A induces obesity through POMC neuron regulation. However, both groups exhibited reduced weight gain and adiposity upon high-fat diet compared to mice carrying only the floxed alleles, suggesting independent effects of Pomc-Cre transgene on body weight. Further analysis reveals that POMC neurons express IL-17A, and reduction in number of POMC neurons in Pomc-Cre mice could be linked to decreased IL-17A expression, which correlates with reduced adipocyte gene expression associated with obesity. Our data underscore an unexpected crosstalk between IL-17A-producing POMC neurons and the endocrine system in obesity regulation.
Collapse
Affiliation(s)
- Rosa Gallo
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Ana Teijeiro
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| |
Collapse
|
7
|
Zhang S, Xie H, Pan P, Wang Q, Yang B, Li Y, Wei Y, Sun Y, Wei Y, Jiang Q, Huang Y. EGCG alleviates heat-stress-induced fat deposition by targeting HSP70 through activation of AMPK-SIRT1-PGC-1α in porcine subcutaneous preadipocytes. Biochem Pharmacol 2024; 225:116250. [PMID: 38705537 DOI: 10.1016/j.bcp.2024.116250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Obesity has emerged as a prominent global health concern, with heat stress posing a significant challenge to both human health and animal well-being. Despite a growing interest in environmental determinants of obesity, very few studies have examined the associations between heat stress-related environmental factors and adiposity. Consequently, there exists a clear need to understand the molecular mechanisms underlying the obesogenic effects of heat stress and to formulate preventive strategies. This study focused on culturing porcine subcutaneous preadipocytes at 41.5 ℃ to induce heat stress, revealing that this stressor triggered apoptosis and fat deposition. Analysis demonstrated an upregulation in the expression of HSP70, BAX, adipogenesis-related genes (PPARγ, AP2, CEBPα and FAS), the p-AMPK/AMPK ratio and SIRT1, PGC-1α in the heat stress group compared to the control group (P < 0.05). Conversely, the expression of lipid lysis-related genes (ATGL, HSL and LPL) and Bcl-2 decreased in the heat stress group compared to the control group (P < 0.05). Furthermore, subsequent activator and/or inhibitor experiments validated that heat stress modulated HSP70 and AMPK signalling pathways to enhance lipogenesis and inhibit lipolysis in porcine subcutaneous preadipocytes. Importantly, this study reveals, for the first time, that EGCG mitigates heat-stress-induced fat deposition by targeting HSP70 through the activation of AMPK-SIRT1-PGC-1α in porcine subcutaneous preadipocytes. These findings elucidate the molecular mechanisms contributing to heat stress-induced obesity and provide a foundation for the potential clinical utilisation of EGCG as a preventive measure against both heat stress and obesity.
Collapse
Affiliation(s)
- Sanbao Zhang
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Hongyue Xie
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China
| | - Peng Pan
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China
| | - Qian Wang
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Bao Yang
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yin Li
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yangyang Wei
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yanjie Sun
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Yirong Wei
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China
| | - Qinyang Jiang
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning Guangxi 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
8
|
Jurisch-Yaksi N, Wachten D, Gopalakrishnan J. The neuronal cilium - a highly diverse and dynamic organelle involved in sensory detection and neuromodulation. Trends Neurosci 2024; 47:383-394. [PMID: 38580512 DOI: 10.1016/j.tins.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
Cilia are fascinating organelles that act as cellular antennae, sensing the cellular environment. Cilia gained significant attention in the late 1990s after their dysfunction was linked to genetic diseases known as ciliopathies. Since then, several breakthrough discoveries have uncovered the mechanisms underlying cilia biogenesis and function. Like most cells in the animal kingdom, neurons also harbor cilia, which are enriched in neuromodulatory receptors. Yet, how neuronal cilia modulate neuronal physiology and animal behavior remains poorly understood. By comparing ciliary biology between the sensory and central nervous systems (CNS), we provide new perspectives on the functions of cilia in brain physiology.
Collapse
Affiliation(s)
- Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Science, Norwegian University of Science and Technology, Erling Skalgssons gate 1, 7491 Trondheim, Norway.
| | - Dagmar Wachten
- Department of Biophysical Imaging, Institute of Innate Immunity, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany; Institute for Human Genetics, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, 07740 Jena, Germany
| |
Collapse
|
9
|
Tomlinson JW. Bardet-Biedl syndrome: A focus on genetics, mechanisms and metabolic dysfunction. Diabetes Obes Metab 2024; 26 Suppl 2:13-24. [PMID: 38302651 DOI: 10.1111/dom.15480] [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: 12/15/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
Bardet-Biedl syndrome (BBS) is a rare, monogenic, multisystem disorder characterized by retinal dystrophy, renal abnormalities, polydactyly, learning disabilities, as well as metabolic dysfunction, including obesity and an increased risk of type 2 diabetes. It is a primary ciliopathy, and causative mutations in more than 25 different genes have been described. Multiple cellular mechanisms contribute to the development of the metabolic phenotype associated with BBS, including hyperphagia as a consequence of altered hypothalamic appetite signalling as well as alterations in adipocyte biology promoting adipocyte proliferation and adipogenesis. Within this review, we describe in detail the metabolic phenotype associated with BBS and discuss the mechanisms that drive its evolution. In addition, we review current approaches to the metabolic management of patients with BBS, including the use of weight loss medications and bariatric surgery. Finally, we evaluate the potential of targeting hypothalamic appetite signalling to limit hyperphagia and induce clinically significant weight loss.
Collapse
Affiliation(s)
- Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, UK
| |
Collapse
|
10
|
Skowronski AA, Leibel RL, LeDuc CA. Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk. Endocr Rev 2024; 45:253-280. [PMID: 37971140 PMCID: PMC10911958 DOI: 10.1210/endrev/bnad031] [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: 06/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS-including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
11
|
Ramakrishnan RK, Srivastava A, Rajan R, Abusnana S, Mussa BM. Investigating the role of an immediate early gene FOS as a potential regulator of autophagic response to hypoglycemia in embryonic hypothalamic neurons. Clin Transl Sci 2024; 17:e13749. [PMID: 38488430 PMCID: PMC10941580 DOI: 10.1111/cts.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/18/2024] Open
Abstract
Hypoglycemia-associated autonomic failure (HAAF) is a well-established complication of diabetes. Although HAAF has serious outcomes such as recurrent morbidity, coma, and death, the mechanisms of HAAF and its pathological components are largely unknown. Our previous studies have revealed that hypoglycemia is associated with the upregulation of an immediate early gene - FOS. In addition, it is documented that glucose deprivation activates neuronal autophagic activities. Therefore, the present study aimed to identify the role of FOS and one of the core components of the autophagy pathway, Beclin-1 (encoded by the BECN1 gene), in the regulation of autophagic mechanisms in embryonic hypothalamic neurons in response to hypoglycemic conditions. Embryonic Mouse Hypothalamic Cell Line N39 (mHypoE-N39 or N39) was cultured in reduced concentrations of glucose (2000, 900, 500, and 200 mg/L). Gene and protein expression, as well as immunofluorescence studies on autophagy were conducted under different reduced glucose concentrations in N39 hypothalamic neurons with and without FOS and BECN1 gene knockdowns (KD). The outcomes of the present study have demonstrated a significant increase in autophagosome formation and subsequent lysosomal degradation in the hypothalamic neurons in response to reduced glucose concentrations. This hypoglycemic response appears to be lowered to a similar extent in the FOS KD and BECN1 KD cells, albeit insignificantly from the negative control, is indicative of the involvement of FOS in the autophagic response of hypothalamic neurons to hypoglycemia. Moreover, the KD cells exhibited a change in morphology and reduced cell viability compared with the control cells. Our findings suggest that reduced FOS expression could potentially be associated with impaired autophagic activities that are dependent on BECN1, which could lead to decreased or blunted hypothalamic activation in response to hypoglycemia, and this, in turn, may contribute to the development of HAAF.
Collapse
Affiliation(s)
- Rakhee K. Ramakrishnan
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Ankita Srivastava
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Reeja Rajan
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Salah Abusnana
- Diabetes and Endocrinology DepartmentUniversity Hospital SharjahSharjahUnited Arab Emirates
- Clinical Science DepartmentCollege of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Bashair M. Mussa
- Basic Medical Science DepartmentCollege of Medicine, University of SharjahSharjahUnited Arab Emirates
| |
Collapse
|
12
|
Hernández-Cáceres MP, Pinto-Nuñez D, Rivera P, Burgos P, Díaz-Castro F, Criollo A, Yañez MJ, Morselli E. Role of lipids in the control of autophagy and primary cilium signaling in neurons. Neural Regen Res 2024; 19:264-271. [PMID: 37488876 PMCID: PMC10503597 DOI: 10.4103/1673-5374.377414] [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: 12/27/2022] [Revised: 03/09/2023] [Accepted: 04/27/2023] [Indexed: 07/26/2023] Open
Abstract
The brain is, after the adipose tissue, the organ with the greatest amount of lipids and diversity in their composition in the human body. In neurons, lipids are involved in signaling pathways controlling autophagy, a lysosome-dependent catabolic process essential for the maintenance of neuronal homeostasis and the function of the primary cilium, a cellular antenna that acts as a communication hub that transfers extracellular signals into intracellular responses required for neurogenesis and brain development. A crosstalk between primary cilia and autophagy has been established; however, its role in the control of neuronal activity and homeostasis is barely known. In this review, we briefly discuss the current knowledge regarding the role of autophagy and the primary cilium in neurons. Then we review the recent literature about specific lipid subclasses in the regulation of autophagy, in the control of primary cilium structure and its dependent cellular signaling in physiological and pathological conditions, specifically focusing on neurons, an area of research that could have major implications in neurodevelopment, energy homeostasis, and neurodegeneration.
Collapse
Affiliation(s)
- María Paz Hernández-Cáceres
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Daniela Pinto-Nuñez
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Patricia Rivera
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Burgos
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Francisco Díaz-Castro
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alfredo Criollo
- Instituto de Investigación en Ciencias Odontológicas (ICOD), Facultad de Odontología, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
| | - Maria Jose Yañez
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Autophagy Research Center, Santiago, Chile
| |
Collapse
|
13
|
Guo DF, Williams PA, Laule C, Seaby C, Zhang Q, Sheffield VC, Rahmouni K. POMC Neuron BBSome Regulation of Body Weight is Independent of its Ciliary Function. FUNCTION 2023; 5:zqad070. [PMID: 38223458 PMCID: PMC10787280 DOI: 10.1093/function/zqad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024] Open
Abstract
The BBSome, a complex of several Bardet-Biedl syndrome (BBS) proteins including BBS1, has emerged as a critical regulator of energy homeostasis. Although the BBSome is best known for its involvement in cilia trafficking, through a process that involve BBS3, it also regulates the localization of cell membrane receptors underlying metabolic regulation. Here, we show that inducible Bbs1 gene deletion selectively in proopiomelanocortin (POMC) neurons cause a gradual increase in body weight, which was associated with higher fat mass. In contrast, inducible deletion of Bbs3 gene in POMC neurons failed to affect body weight and adiposity. Interestingly, loss of BBS1 in POMC neurons led to glucose intolerance and insulin insensitivity, whereas BBS3 deficiency in these neurons is associated with slight impairment in glucose handling, but normal insulin sensitivity. BBS1 deficiency altered the plasma membrane localization of serotonin 5-HT2C receptor (5-HT2CR) and ciliary trafficking of neuropeptide Y2 receptor (NPY2R).In contrast, BBS3 deficiency, which disrupted the ciliary localization of the BBSome, did not interfere with plasma membrane expression of 5-HT2CR, but reduced the trafficking of NPY2R to cilia. We also show that deficiency in BBS1, but not BBS3, alters mitochondria dynamics and decreased total and phosphorylated levels of dynamin-like protein 1 (DRP1) protein. Importantly, rescuing DRP1 activity restored mitochondria dynamics and localization of 5-HT2CR and NPY2R in BBS1-deficient cells. The contrasting effects on energy and glucose homeostasis evoked by POMC neuron deletion of BBS1 versus BBS3 indicate that BBSome regulation of metabolism is not related to its ciliary function in these neurons.
Collapse
Affiliation(s)
- Deng-Fu Guo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Veterans Affairs Health Care System, Iowa City, IA 52242, USA
| | - Paul A Williams
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Connor Laule
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Charles Seaby
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Qihong Zhang
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Veterans Affairs Health Care System, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
He K, Nie Z. System analysis based on the lysosome-related genes identifies HPS4 as a novel therapy target for liver hepatocellular carcinoma. Front Oncol 2023; 13:1221498. [PMID: 37781184 PMCID: PMC10535104 DOI: 10.3389/fonc.2023.1221498] [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: 05/12/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Background Liver cancer is a leading cause of cancer-related deaths worldwide. Lysosomal dysfunction is implicated in cancer progression; however, prognostic prediction models based on lysosome-related genes (LRGs) are lacking in liver cancer. This study aimed to establish an LRG-based model to improve prognosis prediction and explore potential therapeutic targets in liver cancer. Methods Expression profiles of 61 LRGs were analyzed in The Cancer Genome Atlas liver cancer cohorts. There were 14 LRGs identified, and their association with clinical outcomes was evaluated. Unsupervised clustering, Cox regression, and functional assays were performed. Results Patients were classified into high-risk and low-risk subgroups based on the 14 LRGs. The high-risk group had significantly worse overall survival. Aberrant immune infiltration and checkpoint expression were observed in the high-risk group. Furthermore, HPS4 was identified as an independent prognostic indicator. Knockdown of HPS4 suppressed liver cancer cell proliferation and induced apoptosis. Conclusion This study developed an LRG-based prognostic model to improve risk stratification in liver cancer. The potential value of HPS4 as a therapeutic target and biomarker was demonstrated. Regulation of HPS4 may offer novel strategies for precision treatment in liver cancer patients.
Collapse
Affiliation(s)
- Ke‐Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| | - Zhiqiang Nie
- Global Health Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Green TE, Fujita A, Ghaderi N, Heinzen EL, Matsumoto N, Klein KM, Berkovic SF, Hildebrand MS. Brain mosaicism of hedgehog signalling and other cilia genes in hypothalamic hamartoma. Neurobiol Dis 2023; 185:106261. [PMID: 37579995 DOI: 10.1016/j.nbd.2023.106261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023] Open
Abstract
Hypothalamic hamartoma (HH) is a rare benign developmental brain lesion commonly associated with a well characterized epilepsy phenotype. Most individuals with HH are non-syndromic without additional developmental anomalies nor a family history of disease. Nonetheless, HH is a feature of Pallister-Hall (PHS) and Oro-Facial-Digital Type VI (OFD VI) syndromes, both characterized by additional developmental anomalies. Initial genetic of analysis HH began with syndromic HH, where germline inherited or de novo variants in GLI3, encoding a central transcription factor in the sonic hedgehog (Shh) signalling pathway, were identified in most individuals with PHS. Following these discoveries in syndromic HH, the hypothesis that post-zygotic mosaicism in related genes may underly non-syndromic HH was tested. We discuss the identified mosaic variants within individuals with non-syndromic HH, review the analytical methodologies and diagnostic yields, and explore understanding of the functional role of the implicated genes with respect to Shh signalling, and cilia development and function. We also outline future challenges in studying non-syndromic HH and suggest potential novel strategies to interrogate brain mosaicism in HH.
Collapse
Affiliation(s)
- Timothy E Green
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Navid Ghaderi
- Departments of Clinical Neurosciences, Medical Genetics and Community Health Sciences, Hotchkiss Brain Institute & Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Erin L Heinzen
- Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Karl Martin Klein
- Departments of Clinical Neurosciences, Medical Genetics and Community Health Sciences, Hotchkiss Brain Institute & Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Epilepsy Center Frankfurt Rhine-Main and Department of Neurology, Goethe University and University Hospital Frankfurt, Frankfurt am Main, Germany; LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | - Michael S Hildebrand
- Epilepsy Research Centre, Department of Medicine, The University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.
| |
Collapse
|
16
|
Tian JL, Huang CW, Eslami F, Mannino MP, Mai RL, Hart GW. Regulation of Primary Cilium Length by O-GlcNAc during Neuronal Development in a Human Neuron Model. Cells 2023; 12:1520. [PMID: 37296641 PMCID: PMC10252524 DOI: 10.3390/cells12111520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The primary cilium plays critical roles in the homeostasis and development of neurons. Recent studies demonstrate that cilium length is regulated by the metabolic state of cells, as dictated by processes such as glucose flux and O-GlcNAcylation (OGN). The study of cilium length regulation during neuron development, however, has been an area left largely unexplored. This project aims to elucidate the roles of O-GlcNAc in neuronal development through its regulation of the primary cilium. Here, we present findings suggesting that OGN levels negatively regulate cilium length on differentiated cortical neurons derived from human-induced pluripotent stem cells. In neurons, cilium length increased significantly during maturation (after day 35), while OGN levels began to drop. Long-term perturbation of OGN via drugs, which inhibit or promote its cycling, during neuron development also have varying effects. Diminishing OGN levels increases cilium length until day 25, when neural stem cells expand and undergo early neurogenesis, before causing cell cycle exit defects and multinucleation. Elevating OGN levels induces greater primary cilia assembly but ultimately results in the development of premature neurons, which have higher insulin sensitivity. These results indicate that OGN levels and primary cilium length are jointly critical in proper neuron development and function. Understanding the interplays between these two nutrient sensors, O-GlcNAc and the primary cilium, during neuron development is important in paving connections between dysfunctional nutrient-sensing and early neurological disorders.
Collapse
Affiliation(s)
- Jie L. Tian
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Chia-Wei Huang
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Farzad Eslami
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Michael Philip Mannino
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Rebecca Lee Mai
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biology, University of Georgia, Athens, GA 30602, USA
| | - Gerald W. Hart
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA; (C.-W.H.); (F.E.); (M.P.M.); (R.L.M.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
17
|
Donato J. Programming of metabolism by adipokines during development. Nat Rev Endocrinol 2023:10.1038/s41574-023-00828-1. [PMID: 37055548 DOI: 10.1038/s41574-023-00828-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
The intrauterine and early postnatal periods represent key developmental stages in which an organism is highly susceptible to being permanently influenced by maternal factors and nutritional status. Strong evidence indicates that either undernutrition or overnutrition during development can predispose individuals to disease later in life, especially type 2 diabetes mellitus and obesity, a concept known as metabolic programming. Adipose tissue produces important signalling molecules that control energy and glucose homeostasis, including leptin and adiponectin. In addition to their well-characterized metabolic effects in adults, adipokines have been associated with metabolic programming by affecting different aspects of development. Therefore, alterations in the secretion or signalling of adipokines, caused by nutritional insults in early life, might lead to metabolic diseases in adulthood. This Review summarizes and discusses the potential role of several adipokines in inducing metabolic programming through their effects during development. The identification of the endocrine factors that act in early life to permanently influence metabolism represents a key step in understanding the mechanisms behind metabolic programming. Thus, future strategies aiming to prevent and treat these metabolic diseases can be designed, taking into consideration the relationship between adipokines and the developmental origins of health and disease.
Collapse
Affiliation(s)
- Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
18
|
DeMars KM, Ross MR, Starr A, McIntyre JC. Neuronal primary cilia integrate peripheral signals with metabolic drives. Front Physiol 2023; 14:1150232. [PMID: 37064917 PMCID: PMC10090425 DOI: 10.3389/fphys.2023.1150232] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023] Open
Abstract
Neuronal primary cilia have recently emerged as important contributors to the central regulation of energy homeostasis. As non-motile, microtubule-based organelles, primary cilia serve as signaling antennae for metabolic status. The impairment of ciliary structure or function can produce ciliopathies for which obesity is a hallmark phenotype and global ablation of cilia induces non-syndromic adiposity in mouse models. This organelle is not only a hub for metabolic signaling, but also for catecholamine neuromodulation that shapes neuronal circuitry in response to sensory input. The objective of this review is to highlight current research investigating the mechanisms of primary cilium-regulated metabolic drives for maintaining energy homeostasis.
Collapse
Affiliation(s)
- Kelly M. DeMars
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Madeleine R. Ross
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Summer Neuroscience Internship Program, University of Florida, Gainesville, FL, United States
| | - Alana Starr
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
19
|
Wu Y, Zhou J, Yang Y. Peripheral and central control of obesity by primary cilia. J Genet Genomics 2023; 50:295-304. [PMID: 36632916 DOI: 10.1016/j.jgg.2022.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/10/2023]
Abstract
Primary cilia are hair-like structures that protrude from the cell surface. They are capable of sensing external cues and conveying a vast array of signals into cells to regulate a variety of physiological activities. Mutations in cilium-associated genes are linked to a group of diseases with overlapping clinical manifestations, collectively known as ciliopathies. A significant proportion of human ciliopathy cases are accompanied by metabolic disorders such as obesity and type 2 diabetes. Nevertheless, the mechanisms through which dysfunction of primary cilia contributes to obesity are complex. In this article, we present an overview of primary cilia and highlight obesity-related ciliopathies. We also discuss the potential role of primary cilia in peripheral organs, with a focus on adipose tissues. In addition, we emphasize the significance of primary cilia in the central regulation of obesity, especially the involvement of ciliary signaling in the hypothalamic control of feeding behavior. This article therefore proposes a framework of both peripheral and central regulation of obesity by primary cilia, which may benefit further exploration of the ciliary role in metabolic regulation.
Collapse
Affiliation(s)
- Yue Wu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, 250014, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, 250014, China; State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
20
|
Liu S, He Y, Li S, Gao X, Yang F. Kinesin family member 3A induces related diseases via wingless-related integration site/β-catenin signaling pathway. Sci Prog 2023; 106:368504221148340. [PMID: 36594221 PMCID: PMC10358705 DOI: 10.1177/00368504221148340] [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] [Indexed: 01/04/2023]
Abstract
Kinesin family member 3A is an important motor protein that participates in various physiological and pathological processes, including normal tissue development, homeostasis maintenance, tumor infiltration, and migration. The wingless-related integration site/β-catenin signaling pathway is essential for critical molecular mechanisms such as embryonic development, organogenesis, tissue regeneration, and carcinogenesis. Recent studies have examined the molecular mechanisms of kinesin family member 3A, among which the wingless-related integration site/β-catenin signaling pathway has gained attention. The interaction between kinesin family member 3A and the wingless-related integration site/β-catenin signaling pathway is compact and complex but is fascinating and worthy of further study. The upregulation and downregulation of kinesin family member 3A influence many diseases and patient survival through the wingless-related integration site/β-catenin signaling pathway. Therefore, this review mainly focuses on describing the kinesin family member 3A and wingless-related integration site/β-catenin signaling pathways and their associated diseases.
Collapse
Affiliation(s)
- Shupeng Liu
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Yang He
- Clinical Medicine College, North China University of Science and Technology, Tangshan, Hebei province, China
| | - Shifeng Li
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan, Hebei Province, China
| | - Xuemin Gao
- NHC Key Laboratory of Pneumoconiosis, Taiyuan, Shanxi Province, China
| | - Fang Yang
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan, Hebei Province, China
| |
Collapse
|
21
|
Brewer KM, Brewer KK, Richardson NC, Berbari NF. Neuronal cilia in energy homeostasis. Front Cell Dev Biol 2022; 10:1082141. [PMID: 36568981 PMCID: PMC9773564 DOI: 10.3389/fcell.2022.1082141] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022] Open
Abstract
A subset of genetic disorders termed ciliopathies are associated with obesity. The mechanisms behind cilia dysfunction and altered energy homeostasis in these syndromes are complex and likely involve deficits in both development and adult homeostasis. Interestingly, several cilia-associated gene mutations also lead to morbid obesity. While cilia have critical and diverse functions in energy homeostasis, including their roles in centrally mediated food intake and peripheral tissues, many questions remain. Here, we briefly discuss syndromic ciliopathies and monogenic cilia signaling mutations associated with obesity. We then focus on potential ways neuronal cilia regulate energy homeostasis. We discuss the literature around cilia and leptin-melanocortin signaling and changes in ciliary G protein-coupled receptor (GPCR) signaling. We also discuss the different brain regions where cilia are implicated in energy homeostasis and the potential for cilia dysfunction in neural development to contribute to obesity. We close with a short discussion on the challenges and opportunities associated with studies looking at neuronal cilia and energy homeostasis. This review highlights how neuronal cilia-mediated signaling is critical for proper energy homeostasis.
Collapse
Affiliation(s)
- Kathryn M. Brewer
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Katlyn K. Brewer
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Nicholas C. Richardson
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Nicolas F. Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, United States
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
22
|
Angelopoulos I, Gakis G, Birmpas K, Kyrousi C, Habeos EE, Kaplani K, Lygerou Z, Habeos I, Taraviras S. Metabolic regulation of the neural stem cell fate: Unraveling new connections, establishing new concepts. Front Neurosci 2022; 16:1009125. [PMID: 36340763 PMCID: PMC9634649 DOI: 10.3389/fnins.2022.1009125] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
The neural stem cell niche is a key regulator participating in the maintenance, regeneration, and repair of the brain. Within the niche neural stem cells (NSC) generate new neurons throughout life, which is important for tissue homeostasis and brain function. NSCs are regulated by intrinsic and extrinsic factors with cellular metabolism being lately recognized as one of the most important ones, with evidence suggesting that it may serve as a common signal integrator to ensure mammalian brain homeostasis. The aim of this review is to summarize recent insights into how metabolism affects NSC fate decisions in adult neural stem cell niches, with occasional referencing of embryonic neural stem cells when it is deemed necessary. Specifically, we will highlight the implication of mitochondria as crucial regulators of NSC fate decisions and the relationship between metabolism and ependymal cells. The link between primary cilia dysfunction in the region of hypothalamus and metabolic diseases will be examined as well. Lastly, the involvement of metabolic pathways in ependymal cell ciliogenesis and physiology regulation will be discussed.
Collapse
Affiliation(s)
| | - Georgios Gakis
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Kyriakos Birmpas
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Christina Kyrousi
- First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Eginition Hospital, Athens, Greece
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Evagelia Eva Habeos
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Konstantina Kaplani
- Department of Physiology, Medical School, University of Patras, Patras, Greece
| | - Zoi Lygerou
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
| | - Ioannis Habeos
- Division of Endocrinology, Department of Internal Medicine, University of Patras, Patras, Greece
| | - Stavros Taraviras
- Department of Physiology, Medical School, University of Patras, Patras, Greece
- *Correspondence: Stavros Taraviras,
| |
Collapse
|
23
|
Abstract
Cilium formation and regeneration requires new protein synthesis, but the underlying cytosolic translational reprogramming remains largely unknown. Using ribosome footprinting, we performed global translatome profiling during cilia regeneration in Chlamydomonas and uncovered that flagellar genes undergo an early transcriptional activation but late translational repression. This pattern guided our identification of sphingolipid metabolism enzymes, including serine palmitoyltransferase (SPT), as essential regulators for ciliogenesis. Cryo-electron tomography showed that ceramide loss abnormally increased the membrane-axoneme distance and generated bulged cilia. We found that ceramides interact with intraflagellar transport (IFT) particle proteins that IFT motors transport along axoneme microtubules (MTs), suggesting that ceramide-IFT particle-IFT motor-MT interactions connect the ciliary membrane with the axoneme to form rod-shaped cilia. SPT-deficient vertebrate cells were defective in ciliogenesis, and SPT mutations from patients with hereditary sensory neuropathy disrupted cilia, which could be restored by sphingolipid supplementation. These results reveal a conserved role of sphingolipid in cilium formation and link compromised sphingolipid production with ciliopathies.
Collapse
|
24
|
Li Y, Yang S, Liu Y, Qin L, Yang S. IFT20 governs mesenchymal stem cell fate through positively regulating TGF-β-Smad2/3-Glut1 signaling mediated glucose metabolism. Redox Biol 2022; 54:102373. [PMID: 35751983 PMCID: PMC9243161 DOI: 10.1016/j.redox.2022.102373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Aberrant lineage allocation of mesenchymal stem cells (MSCs) could cause bone marrow osteoblast-adipocyte imbalance, and glucose as an important nutrient is required for the maintenance of the MSCs' fate and function. Intraflagellar transport 20 (IFT20) is one of the IFT complex B protein which regulates osteoblast differentiation, and bone formation, but how IFT20 regulates MSCs' fate remains undefined. Here, we demonstrated that IFT20 controls MSC lineage allocation through regulating glucose metabolism during skeletal development. IFT20 deficiency in the early stage of MSCs caused significantly shortened limbs, decreased bone mass and significant increase in marrow fat. However, deletion of IFT20 in the later stage of MSCs and osteocytes just slightly decreased bone mass and bone growth and increased marrow fat. Additionally, we found that loss of IFT20 in MSCs promotes adipocyte formation, which enhances RANKL expression and bone resorption. Conversely, ablation of IFT20 in adipocytes reversed these phenotypes. Mechanistically, loss of IFT20 in MSCs significantly decreased glucose tolerance and suppressed glucose uptake and lactate and ATP production. Moreover, loss of IFT20 significantly decreased the activity of TGF-β-Smad2/3 signaling and reduced the binding activity of Smad2/3 to Glut1 promoter to downregulate Glut1 expression. These findings indicate that IFT20 plays essential roles for preventing MSC lineage allocation into adipocytes through TGF-β-Smad2/3-Glut1 axis.
Collapse
Affiliation(s)
- Yang Li
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shuting Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yang Liu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; The Penn Center for Musculoskeletal Disorders, School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Liu J, Lai F, Hou Y, Zheng R. Leptin signaling and leptin resistance. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:363-384. [PMID: 37724323 PMCID: PMC10388810 DOI: 10.1515/mr-2022-0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/12/2022] [Indexed: 09/20/2023]
Abstract
With the prevalence of obesity and associated comorbidities, studies aimed at revealing mechanisms that regulate energy homeostasis have gained increasing interest. In 1994, the cloning of leptin was a milestone in metabolic research. As an adipocytokine, leptin governs food intake and energy homeostasis through leptin receptors (LepR) in the brain. The failure of increased leptin levels to suppress feeding and elevate energy expenditure is referred to as leptin resistance, which encompasses complex pathophysiological processes. Within the brain, LepR-expressing neurons are distributed in hypothalamus and other brain areas, and each population of the LepR-expressing neurons may mediate particular aspects of leptin effects. In LepR-expressing neurons, the binding of leptin to LepR initiates multiple signaling cascades including janus kinase (JAK)-signal transducers and activators of transcription (STAT) phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT), extracellular regulated protein kinase (ERK), and AMP-activated protein kinase (AMPK) signaling, etc., mediating leptin actions. These findings place leptin at the intersection of metabolic and neuroendocrine regulations, and render leptin a key target for treating obesity and associated comorbidities. This review highlights the main discoveries that shaped the field of leptin for better understanding of the mechanism governing metabolic homeostasis, and guides the development of safe and effective interventions to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Futing Lai
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Yujia Hou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
- Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China
- Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing 100191, China
| |
Collapse
|
26
|
Ávalos Y, Hernández-Cáceres MP, Lagos P, Pinto-Nuñez D, Rivera P, Burgos P, Díaz-Castro F, Joy-Immediato M, Venegas-Zamora L, Lopez-Gallardo E, Kretschmar C, Batista-Gonzalez A, Cifuentes-Araneda F, Toledo-Valenzuela L, Rodriguez-Peña M, Espinoza-Caicedo J, Perez-Leighton C, Bertocchi C, Cerda M, Troncoso R, Parra V, Budini M, Burgos PV, Criollo A, Morselli E. Palmitic acid control of ciliogenesis modulates insulin signaling in hypothalamic neurons through an autophagy-dependent mechanism. Cell Death Dis 2022; 13:659. [PMID: 35902579 PMCID: PMC9334645 DOI: 10.1038/s41419-022-05109-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/21/2023]
Abstract
Palmitic acid (PA) is significantly increased in the hypothalamus of mice, when fed chronically with a high-fat diet (HFD). PA impairs insulin signaling in hypothalamic neurons, by a mechanism dependent on autophagy, a process of lysosomal-mediated degradation of cytoplasmic material. In addition, previous work shows a crosstalk between autophagy and the primary cilium (hereafter cilium), an antenna-like structure on the cell surface that acts as a signaling platform for the cell. Ciliopathies, human diseases characterized by cilia dysfunction, manifest, type 2 diabetes, among other features, suggesting a role of the cilium in insulin signaling. Cilium depletion in hypothalamic pro-opiomelanocortin (POMC) neurons triggers obesity and insulin resistance in mice, the same phenotype as mice deficient in autophagy in POMC neurons. Here we investigated the effect of chronic consumption of HFD on cilia; and our results indicate that chronic feeding with HFD reduces the percentage of cilia in hypothalamic POMC neurons. This effect may be due to an increased amount of PA, as treatment with this saturated fatty acid in vitro reduces the percentage of ciliated cells and cilia length in hypothalamic neurons. Importantly, the same effect of cilia depletion was obtained following chemical and genetic inhibition of autophagy, indicating autophagy is required for ciliogenesis. We further demonstrate a role for the cilium in insulin sensitivity, as cilium loss in hypothalamic neuronal cells disrupts insulin signaling and insulin-dependent glucose uptake, an effect that correlates with the ciliary localization of the insulin receptor (IR). Consistently, increased percentage of ciliated hypothalamic neuronal cells promotes insulin signaling, even when cells are exposed to PA. Altogether, our results indicate that, in hypothalamic neurons, impairment of autophagy, either by PA exposure, chemical or genetic manipulation, cause cilia loss that impairs insulin sensitivity.
Collapse
Affiliation(s)
- Yenniffer Ávalos
- grid.412179.80000 0001 2191 5013Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - María Paz Hernández-Cáceres
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.443909.30000 0004 0385 4466Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile
| | - Pablo Lagos
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Pinto-Nuñez
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Rivera
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Burgos
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Díaz-Castro
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michelle Joy-Immediato
- grid.7870.80000 0001 2157 0406Laboratory for Molecular Mechanics of Cell Adhesion, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leslye Venegas-Zamora
- grid.443909.30000 0004 0385 4466Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Erik Lopez-Gallardo
- grid.443909.30000 0004 0385 4466Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Catalina Kretschmar
- grid.443909.30000 0004 0385 4466Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile
| | - Ana Batista-Gonzalez
- grid.443909.30000 0004 0385 4466Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile
| | - Flavia Cifuentes-Araneda
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lilian Toledo-Valenzuela
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Rodriguez-Peña
- grid.443909.30000 0004 0385 4466Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile
| | - Jasson Espinoza-Caicedo
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudio Perez-Leighton
- grid.7870.80000 0001 2157 0406Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- grid.7870.80000 0001 2157 0406Laboratory for Molecular Mechanics of Cell Adhesion, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Cerda
- grid.443909.30000 0004 0385 4466Integrative Biology Program, Institute of Biomedical Sciences, Facultad de Medicina, Universidad de Chile, Santiago, Chile ,grid.443909.30000 0004 0385 4466Center for Medical Informatics and Telemedicine, Facultad de Medicina, Universidad de Chile, Santiago, Chile ,grid.443909.30000 0004 0385 4466Biomedical Neuroscience Institute, Santiago, Chile
| | - Rodrigo Troncoso
- grid.443909.30000 0004 0385 4466Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile ,grid.443909.30000 0004 0385 4466Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile ,Autophagy Research Center, Santiago, Chile
| | - Valentina Parra
- grid.443909.30000 0004 0385 4466Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile ,Autophagy Research Center, Santiago, Chile ,grid.443909.30000 0004 0385 4466Network for the Study of High-Lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile
| | - Mauricio Budini
- Autophagy Research Center, Santiago, Chile ,grid.443909.30000 0004 0385 4466Laboratory of Molecular and Cellular Pathology, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile
| | - Patricia V. Burgos
- Autophagy Research Center, Santiago, Chile ,grid.442215.40000 0001 2227 4297Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile ,grid.7870.80000 0001 2157 0406Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alfredo Criollo
- grid.443909.30000 0004 0385 4466Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, Universidad de Chile, Santiago, Chile ,grid.443909.30000 0004 0385 4466Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile ,Autophagy Research Center, Santiago, Chile
| | - Eugenia Morselli
- grid.7870.80000 0001 2157 0406Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile ,Autophagy Research Center, Santiago, Chile ,grid.442215.40000 0001 2227 4297Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
27
|
The role of ciliopathy-associated type 3 adenylyl cyclase in infanticidal behavior in virgin adult male mice. iScience 2022; 25:104534. [PMID: 35754726 PMCID: PMC9218507 DOI: 10.1016/j.isci.2022.104534] [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: 10/29/2021] [Revised: 02/11/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Virgin adult male mice often display killing of alien newborns, defined as infanticide, and this behavior is dependent on olfactory signaling. Olfactory perception is achieved by the main olfactory system (MOS) or vomeronasal system (VNS). Although it has been established that the VNS is crucial for infanticide in male mice, the role of the MOS in infanticide remains unknown. Herein, by producing lesions via ZnSO4 perfusion and N-methyl-D-aspartic acid stereotactic injection, we demonstrated that the main olfactory epithelium (MOE), anterior olfactory nucleus (AON), or ventromedial hypothalamus (VMH) is crucial for infanticide in adult males. By using CRISPR-Cas9 coupled with adeno-associated viruses to induce specific knockdown of type 3 adenylyl cyclase (AC3) in these tissues, we further demonstrated that AC3, a ciliopathy-associated protein, in the MOE and the expression of related proteins in the AON or VMH are necessary for infanticidal behavior in virgin adult male mice. MOE lesions and knockdown of AC3 in the MOE result in abnormal infanticidal behavior The infanticidal behavior of male mice is impaired by lesioning of the AON or VMH AC3 knockdown in the AON or VMH affects the infanticidal behavior of male mice
Collapse
|
28
|
Tereshko L, Turrigiano GG, Sengupta P. Primary cilia in the postnatal brain: Subcellular compartments for organizing neuromodulatory signaling. Curr Opin Neurobiol 2022; 74:102533. [PMID: 35405626 PMCID: PMC9167775 DOI: 10.1016/j.conb.2022.102533] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 03/06/2022] [Indexed: 11/03/2022]
Abstract
Primary cilia have well characterized roles in early brain development, relaying signals critical for neurogenesis and brain formation during embryonic stages. Less understood are the contributions of cilia-mediated signaling to postnatal brain function. Several cilia-localized receptors that bind neuropeptides and neurotransmitters endogenous to the brain have been identified in adult neurons, but the functional significance of signaling through these cilia-localized receptors is largely unexplored. Ciliopathic disorders in humans often manifest with neurodevelopmental abnormalities and cognitive deficits. Intriguingly, recent research has also linked several neuropsychiatric disorders and neurodegenerative diseases to ciliary dysfunction. This review summarizes recent evidence suggesting that cilia signaling may dynamically regulate postnatal neuronal physiology and connectivity, and highlights possible links among cilia, neuronal circuitry, neuron survival, and neurological disorders.
Collapse
Affiliation(s)
- Lauren Tereshko
- Department of Biology, Brandeis University, Waltham, MA 02454, USA; Biogen, Cambridge, MA 02142, USA
| | | | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, MA 02454, USA.
| |
Collapse
|
29
|
Croizier S, Bouret SG. Molecular Control of the Development of Hypothalamic Neurons Involved in Metabolic Regulation. J Chem Neuroanat 2022; 123:102117. [DOI: 10.1016/j.jchemneu.2022.102117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/03/2022] [Accepted: 06/03/2022] [Indexed: 10/18/2022]
|
30
|
Lee CH, Kang GM, Kim MS. Mechanisms of Weight Control by Primary Cilia. Mol Cells 2022; 45:169-176. [PMID: 35387896 PMCID: PMC9001153 DOI: 10.14348/molcells.2022.2046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
A primary cilium, a hair-like protrusion of the plasma membrane, is a pivotal organelle for sensing external environmental signals and transducing intracellular signaling. An interesting linkage between cilia and obesity has been revealed by studies of the human genetic ciliopathies Bardet-Biedl syndrome and Alström syndrome, in which obesity is a principal manifestation. Mouse models of cell type-specific cilia dysgenesis have subsequently demonstrated that ciliary defects restricted to specific hypothalamic neurons are sufficient to induce obesity and hyperphagia. A potential mechanism underlying hypothalamic neuron cilia-related obesity is impaired ciliary localization of G protein-coupled receptors involved in the regulation of appetite and energy metabolism. A well-studied example of this is melanocortin 4 receptor (MC4R), mutations in which are the most common cause of human monogenic obesity. In the paraventricular hypothalamus neurons, a blockade of ciliary trafficking of MC4R as well as its downstream ciliary signaling leads to hyperphagia and weight gain. Another potential mechanism is reduced leptin signaling in hypothalamic neurons with defective cilia. Leptin receptors traffic to the periciliary area upon leptin stimulation. Moreover, defects in cilia formation hamper leptin signaling and actions in both developing and differentiated hypothalamic neurons. The list of obesity-linked ciliary proteins is expending and this supports a tight association between cilia and obesity. This article provides a brief review on the mechanism of how ciliary defects in hypothalamic neurons facilitate obesity.
Collapse
Affiliation(s)
- Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon 24252, Korea
| | - Gil Myoung Kang
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
31
|
Bouret SG. Developmental programming of hypothalamic melanocortin circuits. Exp Mol Med 2022; 54:403-413. [PMID: 35474338 PMCID: PMC9076880 DOI: 10.1038/s12276-021-00625-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023] Open
Abstract
The melanocortin system plays a critical role in the central regulation of food intake and energy balance. This system consists of neurons producing pro-opiomelanocortin (POMC), melanocortin receptors (MC4Rs), and the endogenous antagonist agouti-related peptide (AgRP). Pomc and Mc4r deficiency in rodents and humans causes early onset of obesity, whereas a loss of Agrp function is associated with leanness. Accumulating evidence shows that many chronic diseases, including obesity, might originate during early life. The melanocortin system develops during a relatively long period beginning during embryonic life with the birth of POMC and AgRP neurons and continuing postnatally with the assembly of their neuronal circuitry. The development of the melanocortin system requires the tight temporal regulation of molecular factors, such as transcription factors and axon guidance molecules, and cellular mechanisms, such as autophagy. It also involves a complex interplay of endocrine and nutritional factors. The disruption of one or more of these developmental factors can lead to abnormal maturation and function of the melanocortin system and has profound metabolic consequences later in life.
Collapse
Affiliation(s)
- Sebastien G Bouret
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition Research Center, UMR-S 1172, Lille, 59000, France.
- University of Lille, FHU 1,000 Days for Health, Lille, 59000, France.
| |
Collapse
|
32
|
Michelini S, Herbst KL, Precone V, Manara E, Marceddu G, Dautaj A, Maltese PE, Paolacci S, Ceccarini MR, Beccari T, Sorrentino E, Aquilanti B, Velluti V, Matera G, Gagliardi L, Miggiano GAD, Bertelli M. A Multi-Gene Panel to Identify Lipedema-Predisposing Genetic Variants by a Next-Generation Sequencing Strategy. J Pers Med 2022; 12:268. [PMID: 35207755 PMCID: PMC8877075 DOI: 10.3390/jpm12020268] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/25/2022] Open
Abstract
Lipedema is a disabling disease characterized by symmetric enlargement of the lower and/or upper limbs due to deposits of subcutaneous fat, that is easily misdiagnosed. Lipedema can be primary or syndromic, and can be the main feature of phenotypically overlapping disorders. The aim of this study was to design a next-generation sequencing (NGS) panel to help in the diagnosis of lipedema by identifying genes specific for lipedema but also genes for overlapping diseases, and targets for tailored treatments. We developed an NGS gene panel consisting of 305 genes potentially associated with lipedema and putative overlapping diseases relevant to lipedema. The genomes of 162 Italian and American patients with lipedema were sequenced. Twenty-one deleterious variants, according to 3 out of 5 predictors, were detected in PLIN1, LIPE, ALDH18A1, PPARG, GHR, INSR, RYR1, NPC1, POMC, NR0B2, GCKR, PPARA in 17 patients. This extended NGS-based approach has identified a number of gene variants that may be important in the diagnosis of lipedema, that may affect the phenotypic presentation of lipedema or that may cause disorders that could be confused with lipedema. This tool may be important for the diagnosis and treatment of people with pathologic subcutaneous fat tissue accumulation.
Collapse
Affiliation(s)
- Sandro Michelini
- Vascular Diagnostics and Rehabilitation Service, Marino Hospital, ASL Roma 6, 00047 Marino, Italy;
| | - Karen L. Herbst
- Department of Endocrinology and Research, Total Lipedema Care, Los Angeles, CA 90211, USA;
| | - Vincenza Precone
- MAGI EUREGIO, 39100 Bolzano, Italy; (V.P.); (G.M.); (E.S.); (M.B.)
| | - Elena Manara
- MAGI’S LAB, 38068 Rovereto, Italy; (E.M.); (A.D.); (P.E.M.)
| | | | - Astrit Dautaj
- MAGI’S LAB, 38068 Rovereto, Italy; (E.M.); (A.D.); (P.E.M.)
| | | | | | - Maria Rachele Ceccarini
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (M.R.C.); (T.B.)
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, 34148 Trieste, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy; (M.R.C.); (T.B.)
- C.I.B., Consorzio Interuniversitario per le Biotecnologie, 34148 Trieste, Italy
| | - Elisa Sorrentino
- MAGI EUREGIO, 39100 Bolzano, Italy; (V.P.); (G.M.); (E.S.); (M.B.)
| | - Barbara Aquilanti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (B.A.); (V.V.); (G.M.); (L.G.); (G.A.D.M.)
| | - Valeria Velluti
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (B.A.); (V.V.); (G.M.); (L.G.); (G.A.D.M.)
| | - Giuseppina Matera
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (B.A.); (V.V.); (G.M.); (L.G.); (G.A.D.M.)
| | - Lucilla Gagliardi
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (B.A.); (V.V.); (G.M.); (L.G.); (G.A.D.M.)
| | - Giacinto Abele Donato Miggiano
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (B.A.); (V.V.); (G.M.); (L.G.); (G.A.D.M.)
| | - Matteo Bertelli
- MAGI EUREGIO, 39100 Bolzano, Italy; (V.P.); (G.M.); (E.S.); (M.B.)
- MAGI’S LAB, 38068 Rovereto, Italy; (E.M.); (A.D.); (P.E.M.)
| |
Collapse
|
33
|
Skowronski AA, Shaulson ED, Leibel RL, LeDuc CA. The postnatal leptin surge in mice is variable in both time and intensity and reflects nutritional status. Int J Obes (Lond) 2022; 46:39-49. [PMID: 34475504 PMCID: PMC8748198 DOI: 10.1038/s41366-021-00957-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/27/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES The murine postnatal leptin surge occurs within the first 4 weeks of life and is critical for neuronal projection development within hypothalamic feeding circuits. Here we describe the influence of nutritional status on the timing and magnitude of the postnatal leptin surge in mice. METHODS Plasma leptin concentrations were measured 1-3 times per week for the first 4 weeks of life in C57BL/6J pups reared in litters adjusted to 3 (small), 7-8 (normal), or 11-12 (large) pups per dam fed breeder chow or raised in litters of 7-8 by dams fed high-fat diet (HFD) ad libitum starting either prior to conception or at parturition. RESULTS Mice raised in small litters become fatter than pups raised in either normal or large litters. The leptin surge in small litter pups starts earlier, lasts longer, and is dramatically larger in magnitude compared to normal litter pups, even when leptin concentrations are normalized to fat mass. In mice reared in large litters, weight gain is diminished and the surge is both significantly delayed and lower in magnitude compared to control pups. Pups reared by HFD-fed dams (starting preconception or at parturition) are fatter and have augmented leptin surge magnitude compared to pups suckled by chow-fed dams. Surge timing varies depending upon nutritional status of the pup; the source of the surge is primarily subcutaneous adipose tissue. At peak leptin surge, within each group, fat mass and plasma leptin are uncorrelated; in comparison with adults, pups overproduce leptin relative to fat mass. Plasma leptin elevation persists longer than previously described; at postnatal day 27 mice continue overproducing leptin relative to fat mass. CONCLUSIONS In mice, small litter size and maternal HFD feeding during the perinatal period augment the plasma leptin surge whereas large litter size is associated with a delayed surge of reduced magnitude.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Evan D Shaulson
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
34
|
Yang DJ, Hong J, Kim KW. Hypothalamic primary cilium: A hub for metabolic homeostasis. Exp Mol Med 2021; 53:1109-1115. [PMID: 34211092 PMCID: PMC8333261 DOI: 10.1038/s12276-021-00644-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is a global health problem that is associated with adverse consequences such as the development of metabolic disorders, including cardiovascular disease, neurodegenerative disorders, and type 2 diabetes. A major cause of obesity is metabolic imbalance, which results from insufficient physical activity and excess energy intake. Understanding the pathogenesis of obesity, as well as other metabolic disorders, is important in the development of methods for prevention and therapy. The coordination of energy balance takes place in the hypothalamus, a major brain region that maintains body homeostasis. The primary cilium is an organelle that has recently received attention because of its role in controlling energy balance in the hypothalamus. Defects in proteins required for ciliary function and formation, both in humans and in mice, have been shown to cause various metabolic disorders. In this review, we provide an overview of the critical functions of primary cilia, particularly in hypothalamic areas, and briefly summarize the studies on the primary roles of cilia in specific neurons relating to metabolic homeostasis.
Collapse
Affiliation(s)
- Dong Joo Yang
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | | | - Ki Woo Kim
- Departments of Oral Biology and Applied Biological Science, BK21 Four, Yonsei University College of Dentistry, Seoul, 03722, Korea.
| |
Collapse
|
35
|
Wang W, Jack BM, Wang HH, Kavanaugh MA, Maser RL, Tran PV. Intraflagellar Transport Proteins as Regulators of Primary Cilia Length. Front Cell Dev Biol 2021; 9:661350. [PMID: 34095126 PMCID: PMC8170031 DOI: 10.3389/fcell.2021.661350] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Primary cilia are small, antenna-like organelles that detect and transduce chemical and mechanical cues in the extracellular environment, regulating cell behavior and, in turn, tissue development and homeostasis. Primary cilia are assembled via intraflagellar transport (IFT), which traffics protein cargo bidirectionally along a microtubular axoneme. Ranging from 1 to 10 μm long, these organelles typically reach a characteristic length dependent on cell type, likely for optimum fulfillment of their specific roles. The importance of an optimal cilia length is underscored by the findings that perturbation of cilia length can be observed in a number of cilia-related diseases. Thus, elucidating mechanisms of cilia length regulation is important for understanding the pathobiology of ciliary diseases. Since cilia assembly/disassembly regulate cilia length, we review the roles of IFT in processes that affect cilia assembly/disassembly, including ciliary transport of structural and membrane proteins, ectocytosis, and tubulin posttranslational modification. Additionally, since the environment of a cell influences cilia length, we also review the various stimuli encountered by renal epithelia in healthy and diseased states that alter cilia length and IFT.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Brittany M Jack
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Henry H Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Matthew A Kavanaugh
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Robin L Maser
- Department of Clinical Laboratory Sciences, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
36
|
Blaess S, Wachten D. The BBSome: a nexus controlling energy metabolism in the brain. J Clin Invest 2021; 131:148903. [PMID: 33855975 DOI: 10.1172/jci148903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bardet-Biedl syndrome (BBS) is a syndromic ciliopathy that has obesity as a cardinal feature. BBS is caused by mutations in BBS genes. BBS proteins control primary cilia function, and BBS mutations therefore lead to dysfunctional primary cilia. Obesity in patients with BBS is mainly caused by hyperphagia due to dysregulated neuronal function in the brain, in particular in the hypothalamus. However, the mechanism by which mutations in BBS genes result in dysfunction in hypothalamic neurons is not well understood. In this issue of the JCI, Wang et al. used BBS and non-BBS patient-derived induced pluripotent stem cells to generate neurons and hypothalamic neurons. Using this human model system, the authors demonstrated that mutations in BBS genes affected primary cilia function, neuronal morphology, and signaling pathways regulating the function of hypothalamic neurons, which control energy homeostasis. This study provides important insights into the mechanisms of BBS-induced obesity.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology and
| | - Dagmar Wachten
- Institute of Innate Immunity, Department of Biophysical Imaging, Medical Faculty, University of Bonn, Bonn, Germany
| |
Collapse
|
37
|
Tereshko L, Gao Y, Cary BA, Turrigiano GG, Sengupta P. Ciliary neuropeptidergic signaling dynamically regulates excitatory synapses in postnatal neocortical pyramidal neurons. eLife 2021; 10:e65427. [PMID: 33650969 PMCID: PMC7952091 DOI: 10.7554/elife.65427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Primary cilia are compartmentalized sensory organelles present on the majority of neurons in the mammalian brain throughout adulthood. Recent evidence suggests that cilia regulate multiple aspects of neuronal development, including the maintenance of neuronal connectivity. However, whether ciliary signals can dynamically modulate postnatal circuit excitability is unknown. Here we show that acute cell-autonomous knockdown of ciliary signaling rapidly strengthens glutamatergic inputs onto cultured rat neocortical pyramidal neurons and increases spontaneous firing. This increased excitability occurs without changes to passive neuronal properties or intrinsic excitability. Further, the neuropeptide receptor somatostatin receptor 3 (SSTR3) is localized nearly exclusively to excitatory neuron cilia both in vivo and in culture, and pharmacological manipulation of SSTR3 signaling bidirectionally modulates excitatory synaptic inputs onto these neurons. Our results indicate that ciliary neuropeptidergic signaling dynamically modulates excitatory synapses and suggest that defects in this regulation may underlie a subset of behavioral and cognitive disorders associated with ciliopathies.
Collapse
Affiliation(s)
- Lauren Tereshko
- Department of Biology, Brandeis UniversityWalthamUnited States
| | - Ya Gao
- Department of Biology, Brandeis UniversityWalthamUnited States
| | - Brian A Cary
- Department of Biology, Brandeis UniversityWalthamUnited States
| | | | - Piali Sengupta
- Department of Biology, Brandeis UniversityWalthamUnited States
| |
Collapse
|