1
|
Lee TH, Nicolas JC, Quarta C. Molecular and functional mapping of the neuroendocrine hypothalamus: a new era begins. J Endocrinol Invest 2024; 47:2627-2648. [PMID: 38878127 DOI: 10.1007/s40618-024-02411-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/08/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Recent advances in neuroscience tools for single-cell molecular profiling of brain neurons have revealed an enormous spectrum of neuronal subpopulations within the neuroendocrine hypothalamus, highlighting the remarkable molecular and cellular heterogeneity of this brain area. RATIONALE Neuronal diversity in the hypothalamus reflects the high functional plasticity of this brain area, where multiple neuronal populations flexibly integrate a variety of physiological outputs, including energy balance, stress and fertility, through crosstalk mechanisms with peripheral hormones. Intrinsic functional heterogeneity is also observed within classically 'defined' subpopulations of neuroendocrine neurons, including subtypes with distinct neurochemical signatures, spatial organisation and responsiveness to hormonal cues. AIM The aim of this review is to critically evaluate past and current research on the functional diversity of hypothalamic neuroendocrine neurons and their plasticity. It focuses on how this neuronal plasticity in this brain area relates to metabolic control, feeding regulation and interactions with stress and fertility-related neural circuits. CONCLUSION Our analysis provides an original framework for improving our understanding of the hypothalamic regulation of hormone function and the development of neuroendocrine diseases.
Collapse
Affiliation(s)
- T H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - J-C Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - C Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France.
| |
Collapse
|
2
|
Kahale F, Alemi H, Naderi A, Deshpande N, Lee S, Wang S, Singh RB, Dohlman T, Yin J, Jurkunas U, Dana R. Neuropeptide alpha-Melanocyte stimulating hormone preserves corneal endothelial morphology in a murine model of Fuchs dystrophy. Sci Rep 2024; 14:18842. [PMID: 39138334 PMCID: PMC11322312 DOI: 10.1038/s41598-024-69416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Fuchs endothelial corneal dystrophy is a heterogenous disease with multifactorial etiology, and genetic, epigenetic, and exogenous factors contributing to its pathogenesis. DNA damage plays a significant role, with ultraviolet-A (UV-A) emerging as a key contributing factor. We investigate the potential application of neuropeptide α-melanocyte stimulating hormone (α-MSH) in mitigating oxidative stress induced endothelial damage. First, we examined the effects of α-MSH on a cultured human corneal endothelial cell line (HCEnC-21T) exposed to hydrogen peroxide (H2O2) induced oxidative DNA damage. We performed immunofluorescence and flow cytometry to assess DNA damage and cell death in the cultured cells. Additionally, we used an established mouse model that utilizes ultraviolet light to induce corneal endothelial cell damage resulting in decreased CEnC number, increased cell size variability, and decreased percentage of hexagonal cells. This endothelial decompensation leads to an increase in corneal thickness. Following UV-A exposure, the mice were systemically treated with α-MSH, either immediately after exposure (early treatment) or beginning two weeks post-exposure (delayed treatment). To evaluate treatment efficacy, we analyzed CEnC density and morphology using in vivo confocal microscopy, and central corneal thickness using anterior segment optical coherence tomography. Our findings demonstrated that α-MSH treatment effectively protects HCEnC-21T from free-radical induced oxidative DNA damage and subsequent cell death. In vivo, α-MSH treatment, mitigated the loss of CEnC density, deterioration of cell morphology and suppression of the resultant corneal swelling. These results underline the potential application of α-MSH as a therapeutic agent for mitigating corneal endothelial damage.
Collapse
Affiliation(s)
- Francesca Kahale
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Hamid Alemi
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Amirreza Naderi
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Neha Deshpande
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Seokjoo Lee
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Shudan Wang
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Rohan Bir Singh
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Thomas Dohlman
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Jia Yin
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Ula Jurkunas
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA
| | - Reza Dana
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| |
Collapse
|
3
|
Sotelo-Hitschfeld T, Minère M, Klemm P, Borgmann D, Wnuk-Lipinski D, Jais A, Jia X, Corneliussen S, Kloppenburg P, Fenselau H, Brüning JC. GABAergic disinhibition from the BNST to PNOC ARC neurons promotes HFD-induced hyperphagia. Cell Rep 2024; 43:114343. [PMID: 38865247 DOI: 10.1016/j.celrep.2024.114343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/01/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.
Collapse
Affiliation(s)
- Tamara Sotelo-Hitschfeld
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Marielle Minère
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daria Wnuk-Lipinski
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Xianglian Jia
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Svenja Corneliussen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute of Zoology, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Peter Kloppenburg
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Institute of Zoology, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Henning Fenselau
- Policlinic for Endocrinology, Diabetes, and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany; Synaptic Transmission in Energy Homeostasis Research Group, Max Planck Institute for Metabolism Research, Cologne, Germany.
| | - Jens Claus Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany.
| |
Collapse
|
4
|
Haspula D, Cui Z. Neurochemical Basis of Inter-Organ Crosstalk in Health and Obesity: Focus on the Hypothalamus and the Brainstem. Cells 2023; 12:1801. [PMID: 37443835 PMCID: PMC10341274 DOI: 10.3390/cells12131801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Precise neural regulation is required for maintenance of energy homeostasis. Essential to this are the hypothalamic and brainstem nuclei which are located adjacent and supra-adjacent to the circumventricular organs. They comprise multiple distinct neuronal populations which receive inputs not only from other brain regions, but also from circulating signals such as hormones, nutrients, metabolites and postprandial signals. Hence, they are ideally placed to exert a multi-tier control over metabolism. The neuronal sub-populations present in these key metabolically relevant nuclei regulate various facets of energy balance which includes appetite/satiety control, substrate utilization by peripheral organs and glucose homeostasis. In situations of heightened energy demand or excess, they maintain energy homeostasis by restoring the balance between energy intake and expenditure. While research on the metabolic role of the central nervous system has progressed rapidly, the neural circuitry and molecular mechanisms involved in regulating distinct metabolic functions have only gained traction in the last few decades. The focus of this review is to provide an updated summary of the mechanisms by which the various neuronal subpopulations, mainly located in the hypothalamus and the brainstem, regulate key metabolic functions.
Collapse
Affiliation(s)
- Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Zhenzhong Cui
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
| |
Collapse
|
5
|
Wang X, Cong P, Wang X, Wang Z, Liu B, Xue C, Xu J. Docosahexaenoic acid-acylated astaxanthin monoester ameliorates chronic high-fat diet-induced autophagy dysfunction via ULK1 pathway in the hypothalamus of mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:2378-2388. [PMID: 36606564 DOI: 10.1002/jsfa.12429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/21/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Dietary astaxanthin (AST) exhibits the ability to resist lipid accumulation and stimulate hepatic autophagy. Natural AST predominantly exists in stable esterified forms. More importantly, in our previous study, docosahexaenoic acid-acylated AST monoester (AST-DHA) possessed better stability, bioavailability, and neuroprotective ability than AST in free and diester form. However, the AST-DHA mechanisms of action in regulating the obese phenotype and autophagy of the central nervous system remain unclear. RESULTS High-fat diet (HFD)-fed C57BL/6J mice were orally administered AST-DHA (50 mg/kg body weight/d) for 3 days or 8 weeks. AST-DHA supplementation alleviated HFD-induced abnormal body weight gain, significantly enhanced autophagy with an increased microtubule-associated protein light chain 3 II/I (LC3II/I) ratio, and reduced the accumulation of p62/sequestosome 1 (SQSTM1) in the hypothalamus rather than in the hippocampus. Mechanistically, AST-DHA effectively promoted autophagy and autophagosome formation, and most notably rescued the HFD-impaired autophagosome-lysosome fusion (indicated by the colocalization of LC3 and LAMP1) by regulating mTOR- and AMPK-induced phosphorylation of ULK1. Consequently, AST-DHA enhanced hypothalamic autophagy, leading to pro-opiomelanocortin (POMC) cleavage to produce alpha-melanocyte-stimulating hormone (α-MSH). CONCLUSIONS This study identified AST-DHA as an enhancer of autophagy that plays a beneficial role in restoring hypothalamic autophagy, and as a new potential therapeutic agent against HFD-induced obesity. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Peixu Cong
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xincen Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Zhigao Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Bin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
6
|
Hidese S, Yoshida F, Ishida I, Matsuo J, Hattori K, Kunugi H. Plasma neuropeptide levels in patients with schizophrenia, bipolar disorder, or major depressive disorder and healthy controls: A multiplex immunoassay study. Neuropsychopharmacol Rep 2023; 43:57-68. [PMID: 36414415 PMCID: PMC10009433 DOI: 10.1002/npr2.12304] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
AIM We aimed to compare neuropeptide levels between patients with major psychiatric disorders and healthy controls and examine their association with symptoms and cognitive function. METHODS The participants were 149 patients with schizophrenia, 115 patients with bipolar disorder (BD), 186 unremitted patients with major depressive disorder (MDD), and 350 healthy controls. Psychiatric (schizophrenic, manic, and depressive) symptoms, sleep state, and cognitive (premorbid intelligence quotient, general cognitive, and memory) functions were evaluated. A multiplex immunoassay kit was used to measure cerebrospinal fluid (CSF) and plasma α-melanocyte-stimulating hormone (MSH), β-endorphin, neurotensin, oxytocin, and substance P levels. RESULTS The verification assay revealed that CSF α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were too low to be reliably measured, while plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels could be successfully measured. Plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were not significantly different between patients with schizophrenia, BD, or MDD and healthy controls. Plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were not significantly correlated with psychiatric symptom scores in patients with schizophrenia, BD, or MDD and cognitive function scores in patients or healthy controls. CONCLUSION Our data suggest that plasma neuropeptide levels do not elucidate the involvement of neuropeptides in the pathology of schizophrenia, BD, or MDD.
Collapse
Affiliation(s)
- Shinsuke Hidese
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Fuyuko Yoshida
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ikki Ishida
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Junko Matsuo
- Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kotaro Hattori
- Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Hiroshi Kunugi
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| |
Collapse
|
7
|
Puri S, Kenyon BM, Hamrah P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022; 10:1985. [PMID: 36009532 PMCID: PMC9406019 DOI: 10.3390/biomedicines10081985] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/21/2022] Open
Abstract
The transparency of the cornea along with its dense sensory innervation and resident leukocyte populations make it an ideal tissue to study interactions between the nervous and immune systems. The cornea is the most densely innervated tissue of the body and possesses both immune and vascular privilege, in part due to its unique repertoire of resident immune cells. Corneal nerves produce various neuropeptides that have a wide range of functions on immune cells. As research in this area expands, further insights are made into the role of neuropeptides and their immunomodulatory functions in the healthy and diseased cornea. Much remains to be known regarding the details of neuropeptide signaling and how it contributes to pathophysiology, which is likely due to complex interactions among neuropeptides, receptor isoform-specific signaling events, and the inflammatory microenvironment in disease. However, progress in this area has led to an increase in studies that have begun modulating neuropeptide activity for the treatment of corneal diseases with promising results, necessitating the need for a comprehensive review of the literature. This review focuses on the role of neuropeptides in maintaining the homeostasis of the ocular surface, alterations in disease settings, and the possible therapeutic potential of targeting these systems.
Collapse
Affiliation(s)
- Sudan Puri
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Departments of Immunology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
- Cornea Service, Tufts New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
8
|
Zhou L, Wang T, Yu Y, Li M, Sun X, Song W, Wang Y, Zhang C, Fu F. The etiology of poststroke-depression: a hypothesis involving HPA axis. Biomed Pharmacother 2022; 151:113146. [PMID: 35643064 DOI: 10.1016/j.biopha.2022.113146] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022] Open
Abstract
Approximately, one in three ischemic stroke survivors suffered from depression, namely, post-stroke depression (PSD). PSD affects functional rehabilitation and may lead to poor quality of life of patients. There are numerous explanations about the etiologies of PSD. Here, we speculated that PSD are likely to be the result of specific changes in brain pathology. We hypothesized that the stroke-induced hyperactivity of hypothalamic-pituitary-adrenal (HPA) axis plays an important role in PSD. Stroke initiates a complex sequence of events in neuroendocrine system including HPA axis. The HPA axis is involved in the pathophysiology of depression, especially, the overactivity of the HPA axis occurs in major depressive disorder. This review summarizes the possible etiologies of PSD, focusing on the stroke-induced activation of HPA axis, mainly including the stress followed by severe brain damage and the proinflammatory cytokines release. The role of hyperactive of HPA axis in PSD was discussed in detail, which includes the role of high level corticotropin-releasing hormone in PSD, the effects of glucocorticoids on the alterations in specific brain structures, the expression of enzymes, excitotoxicity, the change in intestinal permeability, and the activation of microglia. The relationship between neuroendocrine regulation and inflammation was also described. Finally, the therapy of PSD by regulating HPA axis, neuroendocrine, and immunity was discussed briefly. Nevertheless, the change of HPA axis and the occurring of PSD maybe interact and promote on each other, and future investigations should explore this hypothesis in more depth.
Collapse
Affiliation(s)
- Lin Zhou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yawen Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Mingan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Xiaohui Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Wenhao Song
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Yunjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Ce Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, PR China.
| |
Collapse
|
9
|
Abstract
Pituitary autoimmunity is one of the principal causes of hypopituitarism. Additionally, hypophysitis is one of the immune-related adverse events associated with immunotherapy. Recent case-oriented research has revealed a novel type of autoimmune hypophysitis, anti-PIT-1 hypophysitis, related to isolated adrenocorticotropic hormone (ACTH) deficiency and immune checkpoint inhibitor-related hypophysitis, as a form of paraneoplastic syndrome. Under these conditions, the ectopic expression of pituitary antigens present in tumors evokes a breakdown of immune tolerance, resulting in the production of autoantibodies and autoreactive cytotoxic T cells that specifically harm pituitary cells. Consequently, an innovative clinical entity of paraneoplastic autoimmune hypophysitis has been purported. This novel concept and its underlying mechanisms provide clues for understanding the pathogenesis of autoimmune pituitary diseases and can be applied to other autoimmune diseases. This review discusses the etiology of paraneoplastic autoimmune hypophysitis and its future.
Collapse
Affiliation(s)
- Hironori Bando
- Division of Development of Advanced Therapy for Metabolic Diseases, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Diabetes and Endocrinology, Nara Medical University, Kashihara, Japan.
| |
Collapse
|
10
|
Anju T, Rai NKSR, Kumar A. Sauropus androgynus (L.) Merr.: a multipurpose plant with multiple uses in traditional ethnic culinary and ethnomedicinal preparations. JOURNAL OF ETHNIC FOODS 2022; 9:10. [PMCID: PMC8900104 DOI: 10.1186/s42779-022-00125-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Various plants form the basis of multiple traditional ethnic cuisines and ethnomedicinal practices across the globe. The ethnic cuisines cater to the nutritional, dietary and medicinal requirements of the tribal and rural communities even today. Using literature from various scholarly databases, this study was conducted to consolidate a comprehensive review on the use of Sauropus androgynus (L.) Merr. in various traditional ethnic cuisines and ethnomedicinal preparations across the globe. The survey shows that it is used in multiple ethnic cuisines and is variously known in different countries and among the communities. Further, it possesses multiple nutritional and ethnomedicinal properties. Considering its importance in ethnic foods and ethnomedicinal preparations, it is important to investigate the nutritional composition, phytochemical constitution and pharmacological basis of ethnomedicinal uses. Therefore, we further compiled this information and found that it is a rich source of both micro- and macronutrients and packed with several bioactive compounds. Survey of pharmacological studies on its traditional medicinal uses supports its ethnomedicinal properties. Despite its importance in traditional food and ethnomedicinal systems, it remains underexplored. Limited information on the toxicity of its various extracts shows that further studies should be conducted to understand its safety aspects. Further clinical studies to prospect possible drug candidates from it should be attempted.
Collapse
Affiliation(s)
- Thattantavide Anju
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| | - Nishmitha Kumari S. R. Rai
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| | - Ajay Kumar
- Department of Plant Science, School of Biological Sciences, Central University of Kerala, Periye, Kasaragod, Kerala 671316 India
| |
Collapse
|
11
|
Woszczyło M, Szumny A, Łyczko J, Jezierski T, Krzemińska P, Szczerbal I, Świtoński M, Niżański W, Dzięcioł M. The Case of Atypical Sexual Attractiveness in a Male Domestic Dog-A Case Study. Animals (Basel) 2021; 11:3156. [PMID: 34827888 PMCID: PMC8614462 DOI: 10.3390/ani11113156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
During the ovarian cycle in domestic dogs, females do not accept males during the first days of estrus but become attractive to males from the beginning of proestrus, with this attractiveness persisting until the end of the estrus phase. It is believed that increased estradiol is responsible for the female attractiveness to the males. In this paper we describe the case of strong, but atypical attractiveness of a castrated male to various, adult, intact males, influenced by the emitted semiochemical signals. Any significant changes in the level of hormones typically involved in the process connected with estrus and responsible for sexual arousal in the males were assessed. The case animal was a 4 year old castrated male Border Collie that was extremely attractive to various males, which presented high levels of sexual arousal, with intensive sniffing and licking of the preputial area, specific vocalization, increased salivation and, finally, mating attempts. Clinical examination of the castrated male revealed a lack of testes in the scrotum and abdominal cavity confirmed by USG. Laboratory tests indicated basal levels of estradiol, testosterone, and progesterone (15.23 pg/mL, <0.05 ng/mL, 0.25 ng/mL), and sex was confirmed via cytogenetic and molecular analysis. Chemical analysis (HS-SPME) of the urine indicated a huge similarity to the profile obtained from a bitch in estrus, with an elevated level of acetophenone, which has been previously postulated in the literature as being a characteristic of the estrus phase in female domestic dogs. This case presented very atypical sexual attractiveness, particularly when taking into account the basal levels of hormones which, according to current knowledge, are responsible for the creation of attractiveness. As a hypothesis requiring verification, we propose the idea of involvement of other hormones in the creation of incidental attractiveness or increased production of compounds responsible for attractiveness (sex pheromones) resulting from metabolic events unrelated to reproductive processes. To our knowledge it is the first described case presenting this phenomenon, which, with more detailed study, could shed new light on the process of creation of sexual attraction in the domestic dog.
Collapse
Affiliation(s)
- Martyna Woszczyło
- Department of Reproduction and Clinic of Farm Animals, Wroclaw University of Environmental and Life Sciences, Plac Grunwaldzki 49, 50-366 Wrocław, Poland; (M.W.); (W.N.)
| | - Antoni Szumny
- Department of Chemistry, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (A.S.); (J.Ł.)
| | - Jacek Łyczko
- Department of Chemistry, Wroclaw University of Environmental and Life Sciences, C.K. Norwida 25, 50-375 Wrocław, Poland; (A.S.); (J.Ł.)
| | - Tadeusz Jezierski
- Department of Animal Behaviour and Welfare, Institute of Genetics and Animal Biotechnology of Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland;
| | - Paulina Krzemińska
- Department of Genetics and Animal Breeding, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland; (P.K.); (I.S.); (M.Ś.)
| | - Izabela Szczerbal
- Department of Genetics and Animal Breeding, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland; (P.K.); (I.S.); (M.Ś.)
| | - Marek Świtoński
- Department of Genetics and Animal Breeding, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, Wolynska 33, 60-637 Poznan, Poland; (P.K.); (I.S.); (M.Ś.)
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, Wroclaw University of Environmental and Life Sciences, Plac Grunwaldzki 49, 50-366 Wrocław, Poland; (M.W.); (W.N.)
| | - Michał Dzięcioł
- Department of Reproduction and Clinic of Farm Animals, Wroclaw University of Environmental and Life Sciences, Plac Grunwaldzki 49, 50-366 Wrocław, Poland; (M.W.); (W.N.)
| |
Collapse
|
12
|
Viral Infection Drives the Regulation of Feeding Behavior Related Genes in Salmo salar. Int J Mol Sci 2021; 22:ijms222111391. [PMID: 34768822 PMCID: PMC8583931 DOI: 10.3390/ijms222111391] [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: 07/20/2021] [Revised: 08/26/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
The feeding behavior in fish is a complex activity that relies on the ability of the brain to integrate multiple signals to produce appropriate responses in terms of food intake, energy expenditure, and metabolic activity. Upon stress cues including viral infection or mediators such as the proinflammatory cytokines, prostaglandins, and cortisol, both Pomc and Npy/Agrp neurons from the hypothalamus are stimulated, thus triggering a response that controls both energy storage and expenditure. However, how appetite modulators or neuro-immune cues link pathogenesis and energy homeostasis in fish remains poorly understood. Here, we provide the first evidence of a molecular linkage between inflammation and food intake in Salmon salar. We show that in vivo viral challenge with infectious pancreatic necrosis virus (IPNV) impacts food consumption by activating anorexic genes such as mc4r, crf, and pomcb and 5-HT in the brain of S. salar. At the molecular level, viral infection induces an overall reduction in lipid content in the liver, favoring the production of AA and EPA associated with the increment of elovl2 gene. In addition, infection upregulates leptin signaling and inhibits insulin signaling. These changes are accompanied by a robust inflammatory response represented by the increment of Il-1b, Il-6, Tnfa, and Pge2 as well as an increased cortisol level in vivo. Thus, we propose a model in which hypothalamic neurons respond to inflammatory cytokines and stress-related molecules and interact with appetite induction/inhibition. These findings provide evidence of crosstalk between pathogenesis-driven inflammation and hypothalamic-pituitary-adrenocortical axes in stress-induced food intake behavior in fish.
Collapse
|
13
|
Perissinotti PP, Martínez-Hernández E, He Y, Koob MD, Piedras-Rentería ES. Genetic Deletion of KLHL1 Leads to Hyperexcitability in Hypothalamic POMC Neurons and Lack of Electrical Responses to Leptin. Front Neurosci 2021; 15:718464. [PMID: 34566565 PMCID: PMC8458657 DOI: 10.3389/fnins.2021.718464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Abstract
Kelch-like 1 (KLHL1) is a neuronal actin-binding protein that modulates voltage-gated calcium channels. The KLHL1 knockout (KO) model displays altered calcium channel expression in various brain regions. We analyzed the electrical behavior of hypothalamic POMC (proopiomelanocortin) neurons and their response to leptin. Leptin's effects on POMC neurons include enhanced gene expression, activation of the ERK1/2 pathway and increased electrical excitability. The latter is initiated by activation of the Jak2-PI3K-PLC pathway, which activates TRPC1/5 (Transient Receptor Potential Cation) channels that in turn recruit T-type channel activity resulting in increased excitability. Here we report over-expression of CaV3.1 T-type channels in the hypothalamus of KLHL1 KO mice increased T-type current density and enhanced POMC neuron basal excitability, rendering them electrically unresponsive to leptin. Electrical sensitivity to leptin was restored by partial blockade of T-type channels. The overexpression of hypothalamic T-type channels in POMC neurons may partially contribute to the obese and abnormal feeding phenotypes observed in KLHL1 KO mice.
Collapse
Affiliation(s)
- Paula P Perissinotti
- Cell and Molecular Physiology Department and Neuroscience Division of the Cardiovascular Research Institute, Loyola University Chicago, Maywood, IL, United States
| | - Elizabeth Martínez-Hernández
- Cell and Molecular Physiology Department and Neuroscience Division of the Cardiovascular Research Institute, Loyola University Chicago, Maywood, IL, United States
| | - Yungui He
- Institute for Translational Neuroscience and Department of Lab Medicine & Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Michael D Koob
- Institute for Translational Neuroscience and Department of Lab Medicine & Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Erika S Piedras-Rentería
- Cell and Molecular Physiology Department and Neuroscience Division of the Cardiovascular Research Institute, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
14
|
Gamma-Aminobutyric Acid (GABA) Inhibits α-Melanocyte-Stimulating Hormone-Induced Melanogenesis through GABA A and GABA B Receptors. Int J Mol Sci 2021; 22:ijms22158257. [PMID: 34361022 PMCID: PMC8347673 DOI: 10.3390/ijms22158257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/25/2022] Open
Abstract
Gamma-aminobutyric acid (GABA) is considered the primary inhibitory neurotransmitter in the human cortex. However, whether GABA regulates melanogenesis has not been comprehensively elucidated. In this study, we reveal that GABA (20 mM) significantly inhibited α-melanocyte-stimulating hormone (α-MSH)-induced extracellular (from 354.9% ± 28.4% to 126.5% ± 16.0%) and intracellular melanin contents (from 236.7% ± 11.1% to 102.7% ± 23.1%) in B16F10 melanoma cells, without inducing cytotoxicity. In addition, α-MSH-induced hyperpigmentation in zebrafish larvae was inhibited from 246.3% ± 5.4% to 116.3% ± 3.1% at 40 mM GABA, displaying no apparent cardiotoxicity. We also clarify that the GABA-mediated antimelanogenic properties were related to the direct inhibition of microphthalmia-associated transcription factor (MITF) and tyrosinase expression by inhibiting cyclic adenosine monophosphate (cAMP) and cAMP response element-binding protein (CREB). Furthermore, under α-MSH stimulation, GABA-related antimelanogenic effects were mediated through the GABAA and GABAB receptors, with subsequent inhibition of Ca2+ accumulation. In B16F10 melanoma cells and zebrafish larvae, pretreatment with bicuculline, a GABAA receptor antagonist, and CGP 46381, a GABAB receptor antagonist, reversed the antimelanogenic effect of GABA following α-MSH treatment by upregulating Ca2+ accumulation. In conclusion, our results indicate that GABA inhibits α-MSH-induced melanogenesis. Hence, in addition to the health benefits of GABA in the central nervous system, it could ameliorate hyperpigmentation disorders.
Collapse
|
15
|
Brocato E, Wolstenholme JT. Neuroepigenetic consequences of adolescent ethanol exposure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:45-84. [PMID: 34696879 DOI: 10.1016/bs.irn.2021.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adolescence is a critical developmental period characterized by ongoing brain maturation processes including myelination and synaptic pruning. Adolescents experience heightened reward sensitivity, sensation seeking, impulsivity, and diminished inhibitory self-control, which contribute to increased participation in risky behaviors, including the initiation of alcohol use. Ethanol exposure in adolescence alters memory and cognition, anxiety-like behavior, and ethanol sensitivity as well as brain myelination and dendritic spine morphology, with effects lasting into adulthood. Emerging evidence suggests that epigenetic modifications may explain these lasting effects. Focusing on the amygdala, prefrontal cortex and hippocampus, we review studies investigating the epigenetic consequences of adolescent ethanol exposure. Ethanol metabolism globally increases donor substrates for histone acetylation and histone and DNA methylation, and this chapter discusses how this can further impact epigenetic programming of the adolescent brain. Elucidation of the mechanisms through which ethanol can alter the epigenetic code at specific transcripts may provide therapeutic targets for intervention.
Collapse
Affiliation(s)
- Emily Brocato
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennifer T Wolstenholme
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States; VCU-Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
16
|
Janczy A, Landowska M, Kochan Z. Gut microbiome dysbiosis in anorexia nervosa. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.8601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Anorexia nervosa (AN) is described as an eating disorder, which is characterized by malnutrition,
a fear of gaining body mass, and a disturbed self-body image. This disease is dependent on biological,
psychological and socio-cultural factors. Among the various biological factors, the importance
of intestinal microbiota has recently attracted much attention. Identification of the gut microbiota
dysbiosis in patients with AN has opened new and promising research directions. Recent
observations focus in particular on the association between intestinal microorganisms and the
occurrence of functional gastrointestinal disorders associated with anorexia, anxiety and depression,
as well as the regulation of eating habits. The composition of the gut microbiota differs between
patients with AN and individuals with normal body mass. This is due to the incorrect diet
of patients; on the other hand, there is growing interest in the role of intestinal microbiota in the
pathogenesis of AN, its changes through re-nutrition practices, and in particular the modulation
of intestinal microbiological composition by means of nutritional interventions or the use of preand
probiotics as standard supplements therapy of eating disorders. There is a need for further
research about the microbiome - intestine - brain axis. Furthermore, consequences of changes in
dietary habits as part of AN treatment are also unknown. However, better knowledge about the
relationship between the gut microbiome and the brain can help improve the treatment of this
disorder. This review aims to present the current knowledge about the potential role of intestinal
microbiota in the pathogenesis, course and treatment of AN.
Collapse
Affiliation(s)
- Agata Janczy
- Zakład Biochemii Żywienia, Katedra Żywienia Klinicznego, Gdański Uniwersytet Medyczny
| | - Magdalena Landowska
- Zakład Biochemii Żywienia, Katedra Żywienia Klinicznego, Gdański Uniwersytet Medyczny
| | - Zdzisław Kochan
- Zakład Biochemii Żywienia, Katedra Żywienia Klinicznego, Gdański Uniwersytet Medyczny
| |
Collapse
|
17
|
Castelli M, Georges A, Holleley CE. Corticosterone does not have a role in temperature sex reversal in the central bearded dragon (Pogona vitticeps). JOURNAL OF EXPERIMENTAL ZOOLOGY PART 2021; 335:301-310. [PMID: 33411403 DOI: 10.1002/jez.2441] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 12/18/2020] [Indexed: 12/29/2022]
Abstract
Environmental sex determination (ESD) is common among ectothermic vertebrates. The stress axis and production of stress hormones (corticosteroids) regulates ESD in fish, but evidence of a similar influence in reptiles is sparse and conflicting. The central bearded dragon (Pogona vitticeps) has a system of sex determination involving the interplay between sex chromosomes (ZZ/ZW female heterogamety) and the thermal environment. High egg incubation temperatures induce sex reversal of the ZZ genotype, feminizing chromosomally male individuals. Here we show that corticosterone elevation is not associated with sex reversal in the central bearded dragon, either during embryonic development or adulthood. We also demonstrate experimentally that sex determination is not affected by corticosterone injection into the yolk. This strongly suggests that stress axis upregulation by high temperature during incubation does not cause sex reversal in P. vitticeps. Our work is in general agreement with other research in reptiles, which suggests that the stress axis does not mediate sex in reptiles with ESD. Alternative biological systems may be responsible for capturing environmental conditions during reptile development, such as cellular calcium and redox regulation or the action of temperature-sensitive splicing factors.
Collapse
Affiliation(s)
- Meghan Castelli
- Institute for Applied Ecology, University of Canberra, Canberra, Australia.,Australian National Wildlife Collection, CSIRO, Canberra, Australia
| | - Arthur Georges
- Institute for Applied Ecology, University of Canberra, Canberra, Australia
| | - Clare E Holleley
- Institute for Applied Ecology, University of Canberra, Canberra, Australia.,Australian National Wildlife Collection, CSIRO, Canberra, Australia
| |
Collapse
|
18
|
Liu X, Li H, Cong X, Huo D, Cong L, Wu G. α-MSH-PE38KDEL Kills Melanoma Cells via Modulating Erk1/2/MITF/TYR Signaling in an MC1R-Dependent Manner. Onco Targets Ther 2020; 13:12457-12469. [PMID: 33299329 PMCID: PMC7721307 DOI: 10.2147/ott.s268554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Background/Objective The immunotoxin α-MSH-PE38KDEL consisting of α-MSH and PE38KDEL showed high cytotoxicity on MSH receptor-positive melanoma cells, suggesting that α-MSH-PE38KDEL might be a potent drug for the treatment of melanoma. Herein, we explored whether the Erk1/2/MITF/TYR signaling, a verified target of α-MSH/MC1R, was involved in α-MSH-PE38KDEL-mediated cytotoxicity. Methods Human melanoma cell line A375, mouse melanoma cell line B16-F10, human breast cancer cell line MDA-MB-231 and human primary epidermal melanocytes (HEMa) with different expression levels of MC1R were used in this study. Cell apoptosis and viability were determined by using flow cytometry and MTT assays. Protein expressions were tested by Western blotting. Results The expression levels of MC1R in A375 and B16-F10 cells were significantly higher than that of MDA-MB-231 and HEMa. α-MSH-PE38KDEL treatment induced a significant inhibition in cell viability in A375 and B16-F10 cells, while showed no obvious influence in the viability of MDA-MB-231 and HEMa cells. However, knockdown of MC1R abolished α-MSH-PE38KDEL role in promoting cell apoptosis in A375 and B16-F10 cells, and upregulation of MC1R endowed α-MSH-PE38KDEL function to promote cell apoptosis in MDA-MB-231 and HEMa cells. Additionally, α-MSH-PE38KDEL treatment increased the phosphorylation levels of Erk1/2 and MITF (S73), and decreased MITF and TYR expressions in an MC1R-dependent manner. All of the treatments, including inhibition of Erk1/2 with PD98059, MC1R downregulation and MITF overexpression weakened the anti-tumor role of α-MSH-PE38KDEL in melanoma. Conclusion Collectively, this study indicates that α-MSH-PE38KDEL promotes melanoma cell apoptosis via modulating Erk1/2/MITF/TYR signaling in an MC1R-dependent manner.
Collapse
Affiliation(s)
- Xilin Liu
- Department of Hand Surgery, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Hong Li
- Emergency Medical Department, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Xianling Cong
- Tissue Bank, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Da Huo
- Department of Hand Surgery, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Lele Cong
- Department of Dermatology, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Guangzhi Wu
- Department of Hand Surgery, China Japan Union Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| |
Collapse
|
19
|
Impact of Genetic Variations and Epigenetic Mechanisms on the Risk of Obesity. Int J Mol Sci 2020; 21:ijms21239035. [PMID: 33261141 PMCID: PMC7729759 DOI: 10.3390/ijms21239035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Rare genetic obesity disorders are characterized by mutations of genes strongly involved in the central or peripheral regulation of energy balance. These mutations are effective in causing the early onset of severe obesity and insatiable hunger (hyperphagia), suggesting that the genetic component can contribute to 40–70% of obesity. However, genes’ roles in the processes leading to obesity are still unclear. This review is aimed to summarize the current knowledge of the genetic causes of obesity, especially monogenic obesity, describing the role of epigenetic mechanisms in obesity and metabolic diseases. A comprehensive understanding of the underlying genetic and epigenetic mechanisms, with the metabolic processes they control, will permit adequate management and prevention of obesity.
Collapse
|
20
|
Rau AR, King CM, Hentges ST. Disruption of GABA or glutamate release from POMC neurons in the adult mouse does not affect metabolic end points. Am J Physiol Regul Integr Comp Physiol 2020; 319:R592-R601. [PMID: 32936679 DOI: 10.1152/ajpregu.00180.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proopiomelanocortin (POMC) neurons contribute to the regulation of many physiological processes; the majority of which have been attributed to the release of peptides produced from the POMC prohormone such as α-MSH, which plays key roles in food intake and metabolism. However, it is now clear that POMC neurons also release amino acid transmitters that likely contribute to the overall function of POMC cells. Recent work indicates that constitutive deletion of these transmitters can affect metabolic phenotypes, but also that the expression of GABAergic or glutamatergic markers changes throughout development. The goal of the present study was to determine whether the release of glutamate or GABA from POMC neurons in the adult mouse contributes notably to energy balance regulation. Disturbed release of glutamate or GABA specifically from POMC neurons in adult mice was achieved using a tamoxifen-inducible Cre construct (Pomc-CreERT2) expressed in mice also carrying floxed versions of Slc17a6 (vGlut2) or Gad1 and Gad2, encoding the vesicular glutamate transporter type 2 and GAD67 and GAD65 proteins, respectively. All mice in the experiments received tamoxifen injections, but control mice lacked the tamoxifen-inducible Cre sequence. Body weight was unchanged in Gad1- and Gad2- or vGlut2-deleted female and male mice. Additionally, no significant differences in glucose tolerance or refeeding after an overnight fast were observed. These data collectively suggest that the release of GABA or glutamate from POMC neurons in adult mice does not significantly contribute to the metabolic parameters tested here. In light of prior work, the data also suggest that amino acid transmitter release from POMC cells may contribute to separate functions in the adult versus the developing mouse.
Collapse
Affiliation(s)
- Andrew R Rau
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Connie M King
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
21
|
Skowron K, Kurnik-Łucka M, Dadański E, Bętkowska-Korpała B, Gil K. Backstage of Eating Disorder-About the Biological Mechanisms behind the Symptoms of Anorexia Nervosa. Nutrients 2020; 12:E2604. [PMID: 32867089 PMCID: PMC7551451 DOI: 10.3390/nu12092604] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Anorexia nervosa (AN) represents a disorder with the highest mortality rate among all psychiatric diseases, yet our understanding of its pathophysiological components continues to be fragmentary. This article reviews the current concepts regarding AN pathomechanisms that focus on the main biological aspects involving central and peripheral neurohormonal pathways, endocrine function, as well as the microbiome-gut-brain axis. It emerged from the unique complexity of constantly accumulating new discoveries, which hamper the ability to look at the disease in a more comprehensive way. The emphasis is placed on the mechanisms underlying the main symptoms and potential new directions that require further investigation in clinical settings.
Collapse
Affiliation(s)
- Kamil Skowron
- Department of Pathophysiology, Jagiellonian University Medical College, Czysta St 18, 31-121 Krakow, Poland; (K.S.); (M.K.-Ł.); (E.D.)
| | - Magdalena Kurnik-Łucka
- Department of Pathophysiology, Jagiellonian University Medical College, Czysta St 18, 31-121 Krakow, Poland; (K.S.); (M.K.-Ł.); (E.D.)
| | - Emil Dadański
- Department of Pathophysiology, Jagiellonian University Medical College, Czysta St 18, 31-121 Krakow, Poland; (K.S.); (M.K.-Ł.); (E.D.)
| | - Barbara Bętkowska-Korpała
- Department of Psychiatry, Jagiellonian University Medical College, Institute of Medical Psychology, Jakubowskiego St 2, 30-688 Krakow, Poland;
| | - Krzysztof Gil
- Department of Pathophysiology, Jagiellonian University Medical College, Czysta St 18, 31-121 Krakow, Poland; (K.S.); (M.K.-Ł.); (E.D.)
| |
Collapse
|
22
|
Chaplot K, Jarvela TS, Lindberg I. Secreted Chaperones in Neurodegeneration. Front Aging Neurosci 2020; 12:268. [PMID: 33192447 PMCID: PMC7481362 DOI: 10.3389/fnagi.2020.00268] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis, or proteostasis, is a combination of cellular processes that govern protein quality control, namely, protein translation, folding, processing, and degradation. Disruptions in these processes can lead to protein misfolding and aggregation. Proteostatic disruption can lead to cellular changes such as endoplasmic reticulum or oxidative stress; organelle dysfunction; and, if continued, to cell death. A majority of neurodegenerative diseases involve the pathologic aggregation of proteins that subverts normal neuronal function. While prior reviews of neuronal proteostasis in neurodegenerative processes have focused on cytoplasmic chaperones, there is increasing evidence that chaperones secreted both by neurons and other brain cells in the extracellular - including transsynaptic - space play important roles in neuronal proteostasis. In this review, we will introduce various secreted chaperones involved in neurodegeneration. We begin with clusterin and discuss its identification in various protein aggregates, and the use of increased cerebrospinal fluid (CSF) clusterin as a potential biomarker and as a potential therapeutic. Our next secreted chaperone is progranulin; polymorphisms in this gene represent a known genetic risk factor for frontotemporal lobar degeneration, and progranulin overexpression has been found to be effective in reducing Alzheimer's- and Parkinson's-like neurodegenerative phenotypes in mouse models. We move on to BRICHOS domain-containing proteins, a family of proteins containing highly potent anti-amyloidogenic activity; we summarize studies describing the biochemical mechanisms by which recombinant BRICHOS protein might serve as a therapeutic agent. The next section of the review is devoted to the secreted chaperones 7B2 and proSAAS, small neuronal proteins which are packaged together with neuropeptides and released during synaptic activity. Since proteins can be secreted by both classical secretory and non-classical mechanisms, we also review the small heat shock proteins (sHsps) that can be secreted from the cytoplasm to the extracellular environment and provide evidence for their involvement in extracellular proteostasis and neuroprotection. Our goal in this review focusing on extracellular chaperones in neurodegenerative disease is to summarize the most recent literature relating to neurodegeneration for each secreted chaperone; to identify any common mechanisms; and to point out areas of similarity as well as differences between the secreted chaperones identified to date.
Collapse
Affiliation(s)
| | | | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
23
|
Seo SH, Jo JK, Kim EJ, Park SE, Shin SY, Park KM, Son HS. Metabolomics Reveals the Alteration of Metabolic Pathway by Alpha-Melanocyte-Stimulating Hormone in B16F10 Melanoma Cells. Molecules 2020; 25:molecules25153384. [PMID: 32722640 PMCID: PMC7436294 DOI: 10.3390/molecules25153384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
The purpose of this study was to understand the changes of metabolic pathway induced by alpha-melanocyte-stimulating hormone (α-MSH) in B16F10 melanoma cells in an untargeted metabolomics approach. Cells were treated with 100 nM of α-MSH and then incubated for 48 h. α-MSH increased tyrosinase activity and melanin content by 56.5 and 61.7%, respectively, compared to untreated cells after 48 h of cultivation. The clear separation between groups was observed in the principal component analysis score plot, indicating that the levels of metabolites of melanoma cells were altered by treatment with α-MSH. Metabolic pathways affected by α-MSH were involved in some amino acid metabolisms. The increased levels of fumaric acid, malic acid, oxaloacetic acid and citric acid related to the citric acid cycle pathway after α-MSH treatment suggested enhanced energy metabolism. Metabolic pathways altered by α-MSH treatment can provide useful information to develop new skin pigmentation inhibitors or anti-obesity drugs.
Collapse
Affiliation(s)
- Seung-Ho Seo
- School of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Korea; (S.-H.S.); (J.K.J.); (E.-J.K.); (S.-E.P.)
| | - Jae Kwon Jo
- School of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Korea; (S.-H.S.); (J.K.J.); (E.-J.K.); (S.-E.P.)
| | - Eun-Ju Kim
- School of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Korea; (S.-H.S.); (J.K.J.); (E.-J.K.); (S.-E.P.)
| | - Seong-Eun Park
- School of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Korea; (S.-H.S.); (J.K.J.); (E.-J.K.); (S.-E.P.)
| | - Seo Yeon Shin
- Department of Pharmaceutical Engineering, Dongshin University, Naju, Jeonnam 58245, Korea;
| | - Kyung Mok Park
- Department of Pharmaceutical Engineering, Dongshin University, Naju, Jeonnam 58245, Korea;
- Correspondence: (K.M.P.); (H.-S.S.); Tel.: +82-32-551-3629 (K.M.P.); +82-61-330-3513 (H.-S.S.)
| | - Hong-Seok Son
- School of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Korea; (S.-H.S.); (J.K.J.); (E.-J.K.); (S.-E.P.)
- Correspondence: (K.M.P.); (H.-S.S.); Tel.: +82-32-551-3629 (K.M.P.); +82-61-330-3513 (H.-S.S.)
| |
Collapse
|
24
|
Molecular evolution of the proopiomelanocortin system in Barn owl species. PLoS One 2020; 15:e0231163. [PMID: 32369484 PMCID: PMC7199972 DOI: 10.1371/journal.pone.0231163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/17/2020] [Indexed: 01/20/2023] Open
Abstract
Examination of genetic polymorphisms in outbred wild-living species provides insights into the evolution of complex systems. In higher vertebrates, the proopiomelanocortin (POMC) precursor gives rise to α-, β-, and γ-melanocyte-stimulating hormones (MSH), which are involved in numerous physiological aspects. Genetic defects in POMC are linked to metabolic disorders in humans and animals. In the present study, we undertook an evolutionary genetic approach complemented with biochemistry to investigate the functional consequences of genetic polymorphisms in the POMC system of free-living outbred barn owl species (family Tytonidae) at the molecular level. Our phylogenetic studies revealed a striking correlation between a loss-of-function H9P mutation in the β-MSH receptor-binding motif and an extension of a poly-serine stretch in γ3-MSH to ≥7 residues that arose in the barn owl group 6–8 MYA ago. We found that extension of the poly-serine stretches in the γ-MSH locus affects POMC precursor processing, increasing γ3-MSH production at the expense of γ2-MSH and resulting in an overall reduction of γ-MSH signaling, which may be part of a negative feedback mechanism. Extension of the γ3-MSH poly-serine stretches ≥7 further markedly increases peptide hormone stability in plasma, which is conserved in humans, and is likely relevant to its endocrine function. In sum, our phylogenetic analysis of POMC in wild living owls uncovered a H9P β-MSH mutation subsequent to serine extension in γ3-MSH to 7 residues, which was then followed by further serine extension. The linked MSH mutations highlight the genetic plasticity enabled by the modular design of the POMC gene.
Collapse
|
25
|
Kumar U, Singh S. Role of Somatostatin in the Regulation of Central and Peripheral Factors of Satiety and Obesity. Int J Mol Sci 2020; 21:ijms21072568. [PMID: 32272767 PMCID: PMC7177963 DOI: 10.3390/ijms21072568] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is one of the major social and health problems globally and often associated with various other pathological conditions. In addition to unregulated eating behaviour, circulating peptide-mediated hormonal secretion and signaling pathways play a critical role in food intake induced obesity. Amongst the many peptides involved in the regulation of food-seeking behaviour, somatostatin (SST) is the one which plays a determinant role in the complex process of appetite. SST is involved in the regulation of release and secretion of other peptides, neuronal integrity, and hormonal regulation. Based on past and recent studies, SST might serve as a bridge between central and peripheral tissues with a significant impact on obesity-associated with food intake behaviour and energy expenditure. Here, we present a comprehensive review describing the role of SST in the modulation of multiple central and peripheral signaling molecules. In addition, we highlight recent progress and contribution of SST and its receptors in food-seeking behaviour, obesity (orexigenic), and satiety (anorexigenic) associated pathways and mechanism.
Collapse
|
26
|
Müschen LH, Rhein M, Hoppe V, John N, Schwabe K, Frieling H, Bleich S, Muschler MAN. Alcohol Withdrawal and Proopiomelanocortin Neuropeptides in an Animal Model of Alcohol Dependence. Neuropsychobiology 2020; 78:118-127. [PMID: 31117084 DOI: 10.1159/000499844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/19/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Alcohol is one of the leading threats to health worldwide. Craving for alcohol makes abstinence a difficult challenge by maintaining alcohol dependence. Many studies suppose the hypothalamic-pituitary-adrenal axis, especially the proopiomelanocortin (POMC)-derived neuropeptides, to mediate craving during withdrawal in alcohol dependence. Evidence is available that the two POMC proteins, α-melanocyte-stimulating hormone (α-MSH) and β-endorphin (β-END) are altered by alcohol consumption and influence alcohol consumption, respectively. OBJECTIVES We investigated the dynamics of α-MSH and β-END during alcohol withdrawal and the influence of intraperitoneal administration of either α-MSH or β-END in an established rodent model (Wistar rats) for alcohol dependence. RESULTS After long-term alcohol self-administration over 12 months and repeated deprivation periods for 3 days, we found a significant decrease in α-MSH levels during withdrawal in rodents (p = 0.006) compared to controls, while β-END levels remained unchanged. Treatment with intraperitoneally administered α-MSH and β-END did not affect alcohol drinking behavior after deprivation. CONCLUSION We demonstrate the effects of alcohol deprivation on α-MSH in alcohol-dependent rodents, which appear to mimic α-MSH alteration found after fasting periods during appetite regulation. Therefore, low α-MSH levels are a possible indicator for craving in alcohol-dependent individuals and hence would be a potential target for anti-craving treatment.
Collapse
Affiliation(s)
- Lars Hendrik Müschen
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Departmentof Neurology, Hannover Medical School, Hannover, Germany
| | - Mathias Rhein
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Laboratory for Molecular Neuroscience, Hannover Medical School, Hannover, Germany
| | - Viktoria Hoppe
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Department of Gynecology, Franziskus Hospital, Bielefeld, Germany
| | - Nadine John
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Kerstin Schwabe
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Laboratory for Molecular Neuroscience, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Laboratory for Molecular Neuroscience, Hannover Medical School, Hannover, Germany
| | - Marc André Nicolas Muschler
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany, .,Laboratory for Molecular Neuroscience, Hannover Medical School, Hannover, Germany,
| |
Collapse
|
27
|
Bayer M, Tsiskarishvili N, Stegemann A, Böhm M, König S. Fast oxidation of α-melanocyte-stimulating hormone and derived peptides under laboratory conditions causes irreproducible results-Insights from studies of prolylcarboxypeptidase in human cell types. Pigment Cell Melanoma Res 2019; 33:378-382. [PMID: 31837203 DOI: 10.1111/pcmr.12852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 11/27/2022]
Affiliation(s)
- Malte Bayer
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, University of Münster, Münster, Germany
| | | | - Agatha Stegemann
- Department of Dermatology, University of Münster, Münster, Germany
| | - Markus Böhm
- Department of Dermatology, University of Münster, Münster, Germany
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, University of Münster, Münster, Germany
| |
Collapse
|
28
|
On-off switching of cell cycle and melanogenesis regulation of melanocytes by non-thermal atmospheric pressure plasma-activated medium. Sci Rep 2019; 9:13400. [PMID: 31527659 PMCID: PMC6746696 DOI: 10.1038/s41598-019-50041-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 08/31/2019] [Indexed: 12/28/2022] Open
Abstract
Non-thermal atmospheric pressure (NAP) plasma has demonstrated potential in biomedical applications, such as cancer treatment, bactericidal sterilization, and cell growth promotion or inhibition. In this study, for the first time, we demonstrated on–off switching of cell cycle progression and regulated melanogenesis in normal human skin melanocytes by NAP plasma-activated medium (PAM). The melanocytes were exposed to NAP plasma at durations varying from 0 to 20 min, and the effects of PAM on cell proliferation, cell cycle progression, and melanogenesis were investigated. Although PAM showed no cytotoxicity, the proliferation of melanocytes was inhibited. The melanocyte cell cycle was arrested by PAM for a relatively short period (48 h), after which it recovered slowly. PAM promoted melanogenesis through the activation of the enzymes tyrosinase, tyrosinase-related protein-1, and tyrosinase-related protein-2. These effects seem to be related to reactive oxygen species induced by PAM. Our finding that PAM modulates the cell cycle may provide insight into the recurrence of cancer. The regulation of the melanogenesis of melanocytes may facilitate the control of skin tone without incurring negative side effects.
Collapse
|
29
|
Feeding Stimulates Sphingosine-1-Phosphate Mobilization in Mouse Hypothalamus. Int J Mol Sci 2019; 20:ijms20164008. [PMID: 31426457 PMCID: PMC6720287 DOI: 10.3390/ijms20164008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that the sphingolipid-derived mediator sphingosine-1-phosphate (S1P) reduces food intake by activating G protein-coupled S1P receptor-1 (S1PR1) in the hypothalamus. Here, we examined whether feeding regulates hypothalamic mobilization of S1P and other sphingolipid-derived messengers. We prepared lipid extracts from the hypothalamus of C57Bl6/J male mice subjected to one of four conditions: free feeding, 12 h fasting, and 1 h or 6 h refeeding. Liquid chromatography/tandem mass spectrometry was used to quantify various sphingolipid species, including sphinganine (SA), sphingosine (SO), and their bioactive derivatives SA-1-phosphate (SA1P) and S1P. In parallel experiments, transcription of S1PR1 (encoded in mice by the S1pr1 gene) and of key genes of sphingolipid metabolism (Sptlc2, Lass1, Sphk1, Sphk2) was measured by RT-PCR. Feeding increased levels of S1P (in pmol-mg−1 of wet tissue) and SA1P. This response was accompanied by parallel changes in SA and dihydroceramide (d18:0/18:0), and was partially (SA1P) or completely (S1P) reversed by fasting. No such effects were observed with other sphingolipid species targeted by our analysis. Feeding also increased transcription of Sptlc2, Lass1, Sphk2, and S1pr1. Feeding stimulates mobilization of endogenous S1PR1 agonists S1P and SA1P in mouse hypothalamus, via a mechanism that involves transcriptional up-regulation of de novo sphingolipid biosynthesis. The results support a role for sphingolipid-mediated signaling in the central control of energy balance.
Collapse
|
30
|
α-Melanocyte-Stimulating Hormone Attenuates Neovascularization by Inducing Nitric Oxide Deficiency via MC-Rs/PKA/NF-κB Signaling. Int J Mol Sci 2018; 19:ijms19123823. [PMID: 30513637 PMCID: PMC6321109 DOI: 10.3390/ijms19123823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
α-melanocyte-stimulating hormone (α-MSH) has been characterized as a novel angiogenesis inhibitor. The homeostasis of nitric oxide (NO) plays an important role in neovascularization. However, it remains unclear whether α-MSH mitigates angiogenesis through modulation of NO and its signaling pathway. The present study elucidated the function and mechanism of NO signaling in α-MSH-induced angiogenesis inhibition using cultured human umbilical vein endothelial cells (HUVECs), rat aorta rings, and transgenic zebrafish. By Griess reagent assay, it was found α-MSH dose-dependently reduced the NO release in HUVECs. Immunoblotting and immunofluorescence analysis revealed α-MSH potently suppressed endothelial and inducible nitric oxide synthase (eNOS/iNOS) expression, which was accompanied with inhibition of nuclear factor kappa B (NF-κB) activities. Excessive supply of NO donor l-arginine reversed the α-MSH-induced angiogenesis inhibition in vitro and in vivo. By using antibody neutralization and RNA interference, it was delineated that melanocortin-1 receptor (MC1-R) and melanocortin-2 receptor (MC2-R) participated in α-MSH-induced inhibition of NO production and NF-κB/eNOS/iNOS signaling. This was supported by pharmaceutical inhibition of protein kinase A (PKA), the downstream effector of MC-Rs signaling, using H89 abolished the α-MSH-mediated suppression of NO release and eNOS/iNOS protein level. Therefore, α-MSH exerts anti-angiogenic function by perturbing NO bioavailability and eNOS/iNOS expression in endothelial cells.
Collapse
|
31
|
D'Agostino G, Lyons D, Cristiano C, Lettieri M, Olarte-Sanchez C, Burke LK, Greenwald-Yarnell M, Cansell C, Doslikova B, Georgescu T, Martinez de Morentin PB, Myers MG, Rochford JJ, Heisler LK. Nucleus of the Solitary Tract Serotonin 5-HT 2C Receptors Modulate Food Intake. Cell Metab 2018; 28:619-630.e5. [PMID: 30146485 PMCID: PMC6371983 DOI: 10.1016/j.cmet.2018.07.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/12/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022]
Abstract
To meet the challenge to human health posed by obesity, a better understanding of the regulation of feeding is essential. Medications targeting 5-hydroxytryptamine (5-HT; serotonin) 2C receptors (htr2c; 5-HT2CR) improve obesity. Here we probed the functional significance of 5-HT2CRs specifically within the brainstem nucleus of the solitary tract (5-HT2CRNTS) in feeding behavior. Selective activation of 5-HT2CRNTS decreased feeding and was sufficient to mediate acute food intake reductions elicited by the 5-HT2CR agonist obesity medication lorcaserin. Similar to pro-opiomelanocortin neurons expressed within the hypothalamic arcuate nucleus (POMCARC), a subset of POMCNTS neurons co-expressed 5-HT2CRs and were activated by 5-HT2CR agonists. Knockdown of POMCNTS prevented the acute appetite-suppressive effect of lorcaserin, whereas POMCARC knockdown prevented the full anorectic effect. These data identify 5-HT2CRNTS as a sufficient subpopulation of 5-HT2CRs in reducing food intake when activated and reveal that 5-HT2CR agonist obesity medications require POMC within the NTS and ARC to reduce food intake.
Collapse
Affiliation(s)
- Giuseppe D'Agostino
- Rowett Institute, University of Aberdeen, Aberdeen, UK; Department of Pharmacology, University of Cambridge, Cambridge, UK.
| | - David Lyons
- Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | | | | | | - Luke K Burke
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Megan Greenwald-Yarnell
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | - Martin G Myers
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Lora K Heisler
- Rowett Institute, University of Aberdeen, Aberdeen, UK; Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
32
|
Perron IJ, Keenan BT, Chellappa K, Lahens NF, Yohn NL, Shockley KR, Pack AI, Veasey SC. Dietary challenges differentially affect activity and sleep/wake behavior in mus musculus: Isolating independent associations with diet/energy balance and body weight. PLoS One 2018; 13:e0196743. [PMID: 29746501 PMCID: PMC5945034 DOI: 10.1371/journal.pone.0196743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS Associated with numerous metabolic and behavioral abnormalities, obesity is classified by metrics reliant on body weight (such as body mass index). However, overnutrition is the common cause of obesity, and may independently contribute to these obesity-related abnormalities. Here, we use dietary challenges to parse apart the relative influence of diet and/or energy balance from body weight on various metabolic and behavioral outcomes. MATERIALS AND METHODS Seventy male mice (mus musculus) were subjected to the diet switch feeding paradigm, generating groups with various body weights and energetic imbalances. Spontaneous activity patterns, blood metabolite levels, and unbiased gene expression of the nutrient-sensing ventral hypothalamus (using RNA-sequencing) were measured, and these metrics were compared using standardized multivariate linear regression models. RESULTS Spontaneous activity patterns were negatively related to body weight (p<0.0001) but not diet/energy balance (p = 0.63). Both body weight and diet/energy balance predicted circulating glucose and insulin levels, while body weight alone predicted plasma leptin levels. Regarding gene expression within the ventral hypothalamus, only two genes responded to diet/energy balance (neuropeptide y [npy] and agouti-related peptide [agrp]), while others were related only to body weight. CONCLUSIONS Collectively, these results demonstrate that individual components of obesity-specifically obesogenic diets/energy imbalance and elevated body mass-can have independent effects on metabolic and behavioral outcomes. This work highlights the shortcomings of using body mass-based indices to assess metabolic health, and identifies novel associations between blood biomarkers, neural gene expression, and animal behavior following dietary challenges.
Collapse
Affiliation(s)
- Isaac J. Perron
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: ,
| | - Brendan T. Keenan
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Karthikeyani Chellappa
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nicholas F. Lahens
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nicole L. Yohn
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Keith R. Shockley
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
| | - Allan I. Pack
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sigrid C. Veasey
- Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
33
|
Al-Najim W, le Roux CW, Docherty NG. Integrated insights into the role of alpha-melanocyte stimulatory hormone in the control of food intake and glycaemia. Peptides 2018; 100:243-248. [PMID: 29412826 DOI: 10.1016/j.peptides.2017.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 10/18/2022]
Abstract
Identifying peptide hormones with multipotent actions on both weight and glycaemia can have a significant impact on therapeutic options in the treatment of obesity and diabetes. This has been exemplified by recent advances involving pharmacological exploitation of glucagon-like peptide 1 biology. Herein, we summarise evidence supporting the potential candidacy in this light of alpha-melanocyte stimulatory hormone, an endogenous peptide hormone and a breakdown product of the neuropeptide pro-opiomelanocortin. We reference its well described central actions in the control of food intake and moreover highlight new data pointing to an important role for this peptide hormone in the periphery, in relation to glycaemic control.
Collapse
Affiliation(s)
- Werd Al-Najim
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Investigative Science, Imperial College London, UK
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden; Investigative Science, Imperial College London, UK
| | - Neil G Docherty
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
34
|
Thomas AL, Maekawa F, Kawashima T, Sakamoto H, Sakamoto T, Davis P, Dores RM. Analyzing the effects of co-expression of chick (Gallus gallus) melanocortin receptors with either chick MRAP1 or MRAP2 in CHO cells on sensitivity to ACTH(1-24) or ACTH(1-13)NH 2: Implications for the avian HPA axis and avian melanocortin circuits in the hypothalamus. Gen Comp Endocrinol 2018; 256:50-56. [PMID: 28888694 DOI: 10.1016/j.ygcen.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/29/2017] [Accepted: 09/03/2017] [Indexed: 10/18/2022]
Abstract
In order to better understand the roles that melanocortin receptors (cMCRs) and melanocortin-2 receptor accessory proteins (cMRAP1 and cMRAP2) play in the HPA axis and hypothalamus, adrenal gland and hypothalamus mRNA from 1day-old white leghorn chicks (Gallus gallus), were analyzed by real-time PCR. mRNA was also made for kidney, ovary, and liver. Mrap1 mRNA could be detected in adrenal tissue, but not in any of the other tissues, and mrap2 mRNA was also detected in the adrenal gland. Finally, all five melanocortin receptors mRNAs could be detected in the adrenal gland; mc2r and mc5r mRNAs were the most abundant. To evaluate any potential interactions between MRAP1 and the MCRs that may occur in adrenal cells, individual chick mcr cDNA constructs were transiently expressed in CHO cells either in the presence or absence of a chick mrap1 cDNA, and the transfected cells were stimulated with hACTH(1-24) at concentrations ranging from 10-13M to 10-6M. As expected, MC2R required co-expression with MRAP1 for functional expression; whereas, co-expression of cMC3R with cMRAP1 had no statistically significant effect on sensitivity to hACTH(1-24). However, co-expression of MC4R and MC5R with MRAP1, increased sensitivity for ACTH(1-24) by approximately 35 fold and 365 fold, respectively. However, co-expressing of cMRAP2 with these melanocortin receptors had no effect on sensitivity to hACTH(1-24). Since the real-time PCR analysis detected mrap2 mRNA and mc4r mRNA in the hypothalamus, the interaction between cMC4R and cMRAP2 with respect to sensitivity to ACTH(1-13)NH2 stimulation was also evaluated. However, no effect, either positive or negative, was observed. Finally, the highest levels of mc5r mRNA were detected in liver cells. This observation raises the possibility that in one-day old chicks, activation of the HPA axis may also involve a physiological response from liver cells.
Collapse
|
35
|
Graham TH. Prolylcarboxypeptidase (PrCP) inhibitors and the therapeutic uses thereof: a patent review. Expert Opin Ther Pat 2017; 27:1077-1088. [PMID: 28699813 DOI: 10.1080/13543776.2017.1349104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Prolylcarboxypeptidase (PrCP) is a serine protease that produces or degrades signaling proteins in several important pathways including the renin-angiotensin system (RAS), kallikrein-kinin system (KKS) and pro-opiomelanocortin (POMC) system. PrCP has the potential to be a therapeutic target for cardiovascular, inflammatory and metabolic diseases. Numerous classes of PrCP inhibitors have been developed by rational drug design and from high-throughput screening hits. These inhibitors have been tested in mouse models to assess their potential as new therapeutics. Areas Covered: This review covers the relevant studies that support PrCP as a target for drug discovery. All the significant patent applications and primary literature concerning the development of PrCP inhibitors are discussed. Expert Opinion: The pathways where PrCP is known to operate are complex and many aspects remain to be characterized. Many potent inhibitors of PrCP have been tested in vivo. The variable results obtained from in vivo studies with PrCP inhibitors suggest that additional understanding of the biochemistry and the required therapeutic inhibitor levels is necessary. Additional fundamental research into the signaling pathways is likely required before the true therapeutic potential of PrCP inhibition will be realized.
Collapse
Affiliation(s)
- Thomas H Graham
- a Merck Research Laboratories , Merck & Co., Inc ., Kenilworth , NJ , USA
| |
Collapse
|
36
|
Stijnen P, Ramos-Molina B, O'Rahilly S, Creemers JWM. PCSK1 Mutations and Human Endocrinopathies: From Obesity to Gastrointestinal Disorders. Endocr Rev 2016; 37:347-71. [PMID: 27187081 DOI: 10.1210/er.2015-1117] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prohormone convertase 1/3, encoded by the PCSK1 gene, is a serine endoprotease that is involved in the processing of a variety of proneuropeptides and prohormones. Humans who are homozygous or compound heterozygous for loss-of-function mutations in PCSK1 exhibit a variable and pleiotropic syndrome consisting of some or all of the following: obesity, malabsorptive diarrhea, hypogonadotropic hypogonadism, altered thyroid and adrenal function, and impaired regulation of plasma glucose levels in association with elevated circulating proinsulin-to-insulin ratio. Recently, more common variants in the PCSK1 gene have been found to be associated with alterations in body mass index, increased circulating proinsulin levels, and defects in glucose homeostasis. This review provides an overview of the endocrinopathies and other disorders observed in prohormone convertase 1/3-deficient patients, discusses the possible biochemical basis for these manifestations of the disease, and proposes a model whereby certain missense mutations in PCSK1 may result in proteins with a dominant negative action.
Collapse
Affiliation(s)
- Pieter Stijnen
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Bruno Ramos-Molina
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Stephen O'Rahilly
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - John W M Creemers
- Laboratory for Biochemical Neuroendocrinology (P.S., B.R.-M., J.W.M.C.), Department of Human Genetics, KU Leuven, Leuven 3000, Belgium; and Medical Research Council (MRC) Metabolic Diseases Unit (S.O.), Wellcome Trust-MRC Institute of Metabolic Science, National Institute for Health Research, Cambridge Biomedical Research Centre, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
37
|
Fullerton JN, Gilroy DW. Resolution of inflammation: a new therapeutic frontier. Nat Rev Drug Discov 2016; 15:551-67. [PMID: 27020098 DOI: 10.1038/nrd.2016.39] [Citation(s) in RCA: 585] [Impact Index Per Article: 73.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dysregulated inflammation is a central pathological process in diverse disease states. Traditionally, therapeutic approaches have sought to modulate the pro- or anti-inflammatory limbs of inflammation, with mixed success. However, insight into the pathways by which inflammation is resolved has highlighted novel opportunities to pharmacologically manipulate these processes - a strategy that might represent a complementary (and perhaps even superior) therapeutic approach. This Review discusses the state of the art in the biology of resolution of inflammation, highlighting the opportunities and challenges for translational research in this field.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| |
Collapse
|
38
|
Ramos-Molina B, Martin MG, Lindberg I. PCSK1 Variants and Human Obesity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:47-74. [PMID: 27288825 DOI: 10.1016/bs.pmbts.2015.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PCSK1, encoding prohormone convertase 1/3 (PC1/3), was one of the first genes linked to monogenic early-onset obesity. PC1/3 is a protease involved in the biosynthetic processing of a variety of neuropeptides and prohormones in endocrine tissues. PC1/3 activity is essential for the activating cleavage of many peptide hormone precursors implicated in the regulation of food ingestion, glucose homeostasis, and energy homeostasis, for example, proopiomelanocortin, proinsulin, proglucagon, and proghrelin. A large number of genome-wide association studies in a variety of different populations have now firmly established a link between three PCSK1 polymorphisms frequent in the population and increased risk of obesity. Human subjects with PC1/3 deficiency, a rare autosomal-recessive disorder caused by the presence of loss-of-function mutations in both alleles, are obese and display a complex set of endocrinopathies. Increasing numbers of genetic diagnoses of infants with persistent diarrhea has recently led to the finding of many novel PCSK1 mutations. PCSK1-deficient infants experience severe intestinal malabsorption during the first years of life, requiring controlled nutrition; these children then become hyperphagic, with associated obesity. The biochemical characterization of novel loss-of-function PCSK1 mutations has resulted in the discovery of new pathological mechanisms affecting the cell biology of the endocrine cell beyond simple loss of enzyme activity, for example, dominant-negative effects of certain mutants on wild-type PC1/3 protein, and activation of the cellular unfolded protein response by endoplasmic reticulum-retained mutants. A better understanding of these molecular and cellular pathologies may illuminate possible treatments for the complex endocrinopathy of PCSK1 deficiency, including obesity.
Collapse
Affiliation(s)
- B Ramos-Molina
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - M G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, Los Angeles, CA, United States of America
| | - I Lindberg
- Department of Anatomy and Neurobiology, University of Maryland, Baltimore, MD, United States of America.
| |
Collapse
|
39
|
Antón Palma B, Leff Gelman P, Medecigo Ríos M, Calva Nieves JC, Acevedo Ortuño R, Matus Ortega ME, Hernández Calderón JA, Hernández Miramontes R, Flores Zamora A, Salazar Juárez A. Generation of a novel monoclonal antibody that recognizes the alpha (α)-amidated isoform of a valine residue. BMC Neurosci 2015; 16:65. [PMID: 26463686 PMCID: PMC4603347 DOI: 10.1186/s12868-015-0206-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/01/2015] [Indexed: 11/10/2022] Open
Abstract
Background Alpha (α)-amidation of peptides is a mechanism required for the conversion of prohormones into functional peptide sequences that display biological activities, receptor recognition and signal transduction on target cells. Alpha (α)-amidation occurs in almost all species and amino acids identified in nature. C-terminal valine amide neuropeptides constitute the smallest group of functional peptide compounds identified in neurosecretory structures in vertebrate and invertebrate species. Methods The α-amidated isoform of valine residue (Val-CONH2) was conjugated to KLH-protein carrier and used to immunize mice. Hyperimmune animals displaying high titers of valine amide antisera were used to generate stable hybridoma-secreting mAbs. Three productive hybridoma (P15A4, P17C11, and P18C5) were tested against peptides antigens containing both the C-terminal α-amidated (–CONH2) and free α-carboxylic acid (−COO−) isovariant of the valine residue. Results P18C5 mAb displayed the highest specificity and selectivity against C-terminal valine amidated peptide antigens in different immunoassays. P18C5 mAb-immunoreactivity exhibited a wide distribution along the neuroaxis of the rat brain, particularly in brain areas that did not cross-match with the neuronal distribution of known valine amide neuropeptides (α-MSH, adrenorphin, secretin, UCN1-2). These brain regions varied in the relative amount of putative novel valine amide peptide immunoreactive material (nmol/μg protein) estimated through a fmol-sensitive solid-phase radioimmunoassay (RIA) raised for P18C5 mAb. Conclusions Our results demonstrate the versatility of a single mAb able to differentiate between two structural subdomains of a single amino acid. This mAb offers a wide spectrum of potential applications in research and medicine, whose uses may extend from a biological reagent (used to detect valine amidated peptide substances in fluids and tissues) to a detoxifying reagent (used to neutralize exogenous toxic amide peptide compounds) or as a specific immunoreagent in immunotherapy settings (used to reduce tumor growth and tumorigenesis) among many others.
Collapse
Affiliation(s)
- Benito Antón Palma
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Philippe Leff Gelman
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico. .,Department of Neuroscience, National Institute of Perinatology, Montes Urales # 800, 11000, México D.F., Mexico.
| | - Mayra Medecigo Ríos
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Juan Carlos Calva Nieves
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Rodolfo Acevedo Ortuño
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Maura Epifanía Matus Ortega
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Jorge Alberto Hernández Calderón
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Ricardo Hernández Miramontes
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Anabel Flores Zamora
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| | - Alberto Salazar Juárez
- Molecular Neurobiology and Addictive Neurochemistry Laboratory, National Institute of Psychiatry, Calzada México-Xochimilco #101, 14370, México D.F., Mexico.
| |
Collapse
|
40
|
Cyr NE, Steger JS, Toorie AM, Yang JZ, Stuart R, Nillni EA. Central Sirt1 regulates body weight and energy expenditure along with the POMC-derived peptide α-MSH and the processing enzyme CPE production in diet-induced obese male rats. Endocrinology 2015; 156:961-74. [PMID: 25549049 PMCID: PMC4330311 DOI: 10.1210/en.2014-1970] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of hypothalamic Sirt1 in body weight and energy balance regulation is debated. The first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague-Dawley rats. Central inhibition of Sirt1 decreased body weight and food intake as a result of a forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which in turn increased phosphorylated FoxO1 via improved insulin/phosphorylated AKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-melanocyte-stimulating hormone (α-MSH) maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (triiodothyronine, thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology (N.E.C., J.S.S., A.M.T., J.Z.Y., R.S., E.A.N.), Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island 02903; and Department of Molecular Biology, Cell Biology, and Biochemistry (E.A.N.), Brown University, Providence, Rhode Island 02912
| | | | | | | | | | | |
Collapse
|
41
|
Kim HJ, Hong J, Yoon HJ, Yoon YR, Kim SY. Carboxypeptidase E is a novel modulator of RANKL-induced osteoclast differentiation. Mol Cells 2014; 37:685-90. [PMID: 25220258 PMCID: PMC4179137 DOI: 10.14348/molcells.2014.0179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 12/03/2022] Open
Abstract
Osteoclasts are large polykaryons that have the unique capacity to degrade bone and are generated by the differentiation of myeloid lineage progenitors. To identify the genes involved in osteoclast development, we performed microarray analysis, and we found that carboxypeptidase E (CPE), a prohormone processing enzyme, was highly upregulated in osteoclasts compared with their precursors, bone marrow-derived macrophages (BMMs). Here, we demonstrate a novel role for CPE in receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation. The overexpression of CPE in BMMs increases the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts and the expression of c-Fos and nuclear factor of activated T cells c1 (NFATc1), which are key regulators in osteoclastogenesis. Furthermore, employing CPE knockout mice, we show that CPE deficiency attenuates osteoclast formation. Together, our data suggest that CPE might be an important modulator of RANKL-induced osteoclast differentiation.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, Clinical Trial Center, School of Medicine, Kyungpook National University, Daegu 700-422, Korea
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu 700-412, Korea
| | - JungMin Hong
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu 700-412, Korea
| | - Hye-Jin Yoon
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu 700-412, Korea
| | - Young-Ran Yoon
- Department of Molecular Medicine, Cell and Matrix Research Institute, Clinical Trial Center, School of Medicine, Kyungpook National University, Daegu 700-422, Korea
| | - Shin-Yoon Kim
- Skeletal Diseases Genome Research Center, Kyungpook National University Hospital, Daegu 700-412, Korea
- Department of Orthopedic Surgery, School of Medicine, Kyungpook National University, Daegu 700-422, Korea
| |
Collapse
|
42
|
Cyr NE, Steger JS, Toorie AM, Yang JZ, Stuart R, Nillni EA. Central Sirt1 regulates body weight and energy expenditure along with the POMC-derived peptide α-MSH and the processing enzyme CPE production in diet-induced obese male rats. Endocrinology 2014; 155:2423-35. [PMID: 24773342 PMCID: PMC4060185 DOI: 10.1210/en.2013-1998] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the periphery, the nutrient-sensing enzyme Sirtuin 1 (silent mating type information regulation 2 homolog 1 [Sirt1]) reduces body weight in diet-induced obese (DIO) rodents. However, the role of Sirt1 in the brain, particularly the hypothalamus, in body weight and energy balance regulation is debated. Among the first studies to reveal that central Sirt1 regulates body weight came from experiments in our laboratory using Sprague Dawley rats. In that study, central inhibition of Sirt1 decreased body weight and food intake as a result of a Forkhead box protein O1 (FoxO1)-mediated increase in the anorexigenic proopiomelanocortin (POMC) and decrease in the orexigenic Agouti-related peptide in the hypothalamic arcuate nucleus. Here, we demonstrate that central inhibition of Sirt1 in DIO decreased body weight and increased energy expenditure at higher levels as compared with the lean counterpart. Brain Sirt1 inhibition in DIO increased acetylated FoxO1, which, in turn, increased phosphorylated FoxO1 via improved insulin/pAKT signaling. Elevated acetylated FoxO1 and phosphorylated FoxO1 increased POMC along with the α-MSH maturation enzyme carboxypeptidase E, which resulted in more of the bioactive POMC product α-MSH released into the paraventricular nucleus. Increased in α-MSH led to augmented TRH levels and circulating T3 levels (thyroid hormone). These results indicate that inhibiting hypothalamic Sirt1 in DIO enhances the activity of the hypothalamic-pituitary-thyroid axis, which stimulates energy expenditure. Because we show that blocking central Sirt1 causes physiological changes that promote a negative energy balance in an obese individual, our results support brain Sirt1 as a significant target for weight loss therapeutics.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology (N.E.C, J.S.S., A.M.T., J.Z.Y, R.S., E.A.N.), Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island 02903; and Department of Molecular Biology, Cell Biology and Biochemistry (E.A.N.), Brown University, Providence, Rhode Island 02912
| | | | | | | | | | | |
Collapse
|
43
|
Fang J, Han D, Hong J, Zhang H, Ying Y, Tian Y, Zhang L, Lin J. SVα-MSH, a novel α-melanocyte stimulating hormone analog, ameliorates autoimmune encephalomyelitis through inhibiting autoreactive CD4(+) T cells activation. J Neuroimmunol 2014; 269:9-19. [PMID: 24518673 DOI: 10.1016/j.jneuroim.2014.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/18/2014] [Accepted: 01/21/2014] [Indexed: 10/25/2022]
Abstract
Alpha-melanocyte stimulating hormone (α-MSH) plays a crucial role in the regulation of immune and inflammatory reactions. Here we report that SVα-MSH, a novel α-MSH analog, could ameliorate the clinical severity of experimental autoimmune encephalomyelitis (EAE) in a preventive and therapeutic manner. SVα-MSH treatment induced the production of regulatory T (Treg) cells and reduced the Th17 cells in the CNS of EAE mice. SVα-MSH-treated PLP peptide 139-151-specific T cells showed a down-regulation of T cell activation markers CD69 and CD134. SVα-MSH did not induce apoptosis but blocked the G1/S phase transition, reduced the expression of cyclin E, Cdk2 and the activity of NFAT and AP-1 transcription factors. Thus, SVα-MSH acts as a novel immunotherapeutic approach in the treatment of autoimmune attack on the CNS.
Collapse
Affiliation(s)
- Jie Fang
- Department of Dermatology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Deping Han
- Department of Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Jinsheng Hong
- Department of Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Hengshan Zhang
- Department of Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Ying Ying
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Yeping Tian
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Lurong Zhang
- Department of Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Jianhua Lin
- Department of Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, China
| |
Collapse
|
44
|
Taylor AW. Alpha-melanocyte stimulating hormone (α-MSH) is a post-caspase suppressor of apoptosis in RAW 264.7 macrophages. PLoS One 2013; 8:e74488. [PMID: 24009773 PMCID: PMC3757010 DOI: 10.1371/journal.pone.0074488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/01/2013] [Indexed: 01/04/2023] Open
Abstract
The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) is an important regulator of immune cell activity within the immunosuppressive ocular microenvironment. Its constitutive presence not only suppresses macrophage inflammatory activity, it also participates in retinal pigment epithelial cell (RPE) mediated activation of macrophages to function similar to myeloid suppressor cells. In addition, α-MSH promotes survival of the alternatively activated macrophages where without α-MSH RPE induce apoptosis in the macrophages, which is seen as increased TUNEL stained cells. Since there is little know about α-MSH as an anti-apoptotic factor, the effects of α-MSH on caspase activity, mitochondrial membrane potential, Bcl2 to BAX expression, along with TUNEL staining, and Annexin V binding were examined in RAW 264.7 macrophages under serum-starved conditions that trigger apoptosis. There was no effect of α-MSH on activated Caspase 9 and Caspase 3 while there was suppression of Caspase 8 activity. In addition, α-MSH did not improve mitochondrial membrane potential, change the ratio between Bcl-2 and BAX, nor reduce Annexin V binding. These results demonstrate that the diminution in TUNEL staining by α-MSH is through α-MSH mediating suppression of the apoptotic pathway that is post-Caspase 3, but before DNA fragmentation. Therefore, as α-MSH promotes the alternative activation of macrophages it also provides a survival signal, and the potential for the caspases to participate in non-apoptotic activities that can contribute to an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Andrew W Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
45
|
Cyr NE, Toorie AM, Steger JS, Sochat MM, Hyner S, Perello M, Stuart R, Nillni EA. Mechanisms by which the orexigen NPY regulates anorexigenic α-MSH and TRH. Am J Physiol Endocrinol Metab 2013; 304:E640-50. [PMID: 23321476 PMCID: PMC3602689 DOI: 10.1152/ajpendo.00448.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 01/12/2013] [Indexed: 01/24/2023]
Abstract
Protein posttranslational processing is a cellular mechanism fundamental to the generation of bioactive peptides, including the anorectic α-melanocyte-stimulating hormone (α-MSH) and thyrotropin-releasing hormone (TRH) peptides produced in the hypothalamic arcuate (ARC) and paraventricular (PVN) nuclei, respectively. Neuropeptide Y (NPY) promotes positive energy balance in part by suppressing α-MSH and TRH. The mechanism by which NPY regulates α-MSH output, however, is not well understood. Our results reveal that NPY inhibited the posttranslational processing of α-MSH's inactive precursor proopiomelanocortin (POMC) by decreasing the prohormone convertase-2 (PC2). We also found that early growth response protein-1 (Egr-1) and NPY-Y1 receptors mediated the NPY-induced decrease in PC2. NPY given intra-PVN also decreased PC2 in PVN samples, suggesting a reduction in PC2-mediated pro-TRH processing. In addition, NPY attenuated the α-MSH-induced increase in TRH production by two mechanisms. First, NPY decreased α-MSH-induced CREB phosphorylation, which normally enhances TRH transcription. Second, NPY decreased the amount of α-MSH in the PVN. Collectively, these results underscore the significance of the interaction between NPY and α-MSH in the central regulation of energy balance and indicate that posttranslational processing is a mechanism that plays a specific role in this interaction.
Collapse
Affiliation(s)
- Nicole E Cyr
- Division of Endocrinology, Department of Medicine, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu GS, Peshavariya H, Higuchi M, Brewer AC, Chang CWT, Chan EC, Dusting GJ. Microphthalmia-associated transcription factor modulates expression of NADPH oxidase type 4: a negative regulator of melanogenesis. Free Radic Biol Med 2012; 52:1835-43. [PMID: 22401855 DOI: 10.1016/j.freeradbiomed.2012.02.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 02/23/2012] [Accepted: 02/24/2012] [Indexed: 12/13/2022]
Abstract
How signaling via reactive oxygen species (ROS) influences skin pigmentation is unclear. We have investigated how NADPH oxidase-derived ROS modulates the expression of the key pigment "melanin" synthesizing enzymes in B16 mouse melanoma cells. A melanin inducer α-melanocyte-stimulating hormone (α-MSH) caused ROS generation that was inhibited by the NADPH oxidase inhibitor Diphenyleneiodonium (DPI) and was insensitive to antagonists of other ROS-producing enzyme systems including mitochondrial enzymes, cycloxygenase, and xanthine oxidase. NADPH oxidase 4 (Nox4) was found to be the most abundant isoform expressed in B16 cells, and its gene levels, as well as ROS generation, were enhanced by α-MSH. Interestingly, silencing Nox4 gene expression with Nox4 siRNA augmented melanin formation under basal conditions and after α-MSH stimulation, demonstrating that constitutive or stimulated Nox4-dependent ROS inhibits melanin formation. This process may be mediated by targeting the promoter region of a melanin synthesizing enzyme tyrosinase, because Nox4 siRNA enhanced tyrosinase promoter activity. Moreover, inhibition of tyrosinase mRNA expression in Nox4 siRNA-treated cells by blocking de novo mRNA and protein synthesis with actinomycin D and cycloheximide respectively indicates that Nox4 repression induces melanogenesis by increasing tyrosinase gene expression. We also found that α-MSH activated its downstream signal transducer microphthalmia-associated transcription factor (MITF) to stimulate Nox4 gene expression. We thus identified a novel mechanism by MITF signaling that in turn stimulates Nox4 to drive ROS generation, thereby repressing melanin synthesis. Such sequence of actions appears to act as an internal feedback mechanism to fine-tune melanin synthesis in response to exogenous challenges such as UV radiation.
Collapse
Affiliation(s)
- Guei-Sheung Liu
- O'Brien Institute, University of Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
47
|
Kaushik S, Arias E, Kwon H, Lopez NM, Athonvarangkul D, Sahu S, Schwartz GJ, Pessin JE, Singh R. Loss of autophagy in hypothalamic POMC neurons impairs lipolysis. EMBO Rep 2012; 13:258-65. [PMID: 22249165 DOI: 10.1038/embor.2011.260] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 12/16/2011] [Accepted: 12/16/2011] [Indexed: 11/09/2022] Open
Abstract
Autophagy degrades cytoplasmic contents to achieve cellular homeostasis. We show that selective loss of autophagy in hypothalamic proopiomelanocortin (POMC) neurons decreases α-melanocyte-stimulating hormone (MSH) levels, promoting adiposity, impairing lipolysis and altering glucose homeostasis. Ageing reduces hypothalamic autophagy and α-MSH levels, and aged-mice phenocopy, the adiposity and lipolytic defect observed in POMC neuron autophagy-null mice. Intraperitoneal isoproterenol restores lipolysis in both models, demonstrating normal adipocyte catecholamine responsiveness. We propose that an unconventional, autophagosome-mediated form of secretion in POMC neurons controls energy balance by regulating α-MSH production. Modulating hypothalamic autophagy might have implications for preventing obesity and metabolic syndrome of ageing.
Collapse
Affiliation(s)
- Susmita Kaushik
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rabey FM, Gadepalli R, Diano S, Cheng Q, Tabrizian T, Gailani D, Rimoldi J, Shariat-Madar Z. Influence of a novel inhibitor (UM8190) of prolylcarboxypeptidase (PRCP) on appetite and thrombosis. Curr Med Chem 2012; 19:4194-206. [PMID: 22664251 PMCID: PMC4367813 DOI: 10.2174/092986712802430036] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 04/02/2012] [Accepted: 04/02/2012] [Indexed: 11/22/2022]
Abstract
Preclinical pharmacological characterization of a novel inhibitor (UM8190) of prolylcarboxypeptidase (PRCP) was investigated. We synthesized and evaluated a library of proline-based analogs as prospective recombinant PRCP (rPRCP) inhibitors and inhibitors of PRCP-dependent prekallikrein (PK) activation on human pulmonary artery endothelial cells (HPAEC). Among the newly synthesized compounds, UM8190 was further characterized in vivo using methods that encompassed a mouse carotid artery thrombosis model and animal model of food consumption. (S)-N-dodecyl-1-((S)-pyrrolidine-2-carbonyl) pyrrolidine-2-carboxamide [Compound 3 (UM8190)] was selected for further evaluation from the initial assessment of its PRCP inhibitory action (K(i)= 43 μM) coupled with its ability to block PRCP-dependent PK activation on HPAEC (K(i)= 34 μM). UM8190 demonstrated excellent selectivity against a panel of carboxypeptidases and serine proteases and blocked bradykinin (BK) generation and BK-induced permeability by 100%, suggesting that it may be useful in preventing the local production of large amounts of BK. Furthermore, UM8190 showed an anorexigenic effect when systemically administered to fasted mice, reducing food intake in a dose- and time-dependent manner. In a mouse carotid artery thrombosis model, it also demonstrated an antithrombotic effect. UM8190 is a selective PRCP inhibitor and it may represent a new anorexigenic, and antithrombotic drug, that works by inhibiting PRCP-mediated mechanisms.
Collapse
Affiliation(s)
- F. M. Rabey
- School of Pharmacy, Department of Pharmacology
| | - R.S.V.S. Gadepalli
- Department of Medicinal Chemistry, University of Mississippi, University, MS 38677-1848
| | - S. Diano
- Program in Cell Signaling and Neurobiology of Metabolism, Departments of Obstetrics, Gynecology & Reproductive Sciences, Neurobiology and Comparative Medicine Yale University School of Medicine, New Haven, CT 06510, USA
| | - Q. Cheng
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232-6307
| | | | - D. Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232-6307
| | - J.M. Rimoldi
- Department of Medicinal Chemistry, University of Mississippi, University, MS 38677-1848
| | | |
Collapse
|
49
|
Zierath D, Tanzi P, Cain K, Shibata D, Becker K. Plasma α-melanocyte stimulating hormone predicts outcome in ischemic stroke. Stroke 2011; 42:3415-20. [PMID: 21960572 DOI: 10.1161/strokeaha.111.627331] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE α-Melanocyte stimulating hormone (α-MSH) is an endogenously produced neuropeptide derived from the same precursor as adrenocorticotropic hormone. α-MSH has profound immunomodulatory properties and may also be neuroprotective. Nothing is known about α-MSH and changes in its plasma concentrations in patients with acute ischemic stroke. METHODS In this prospective observational study, plasma concentrations of α-MSH, adrenocorticotropic hormone, cortisol, and interleukin 6 were assessed longitudinally over the course of 1 year after stroke onset in 111 patients. Logistic regression was used to the effect of initial plasma α-MSH, adrenocorticotropic hormone, cortisol, and interleukin 6 on long-term outcome. RESULTS There was an early decrease in plasma α-MSH in patients with severe stroke (National Institutes of Health Stroke Scale≥17) that normalized over the course of the year; these same patients evidenced elevations in plasma cortisol and interleukin 6. Higher initial plasma α-MSH, but not adrenocorticotropic hormone, cortisol, or interleukin 6, was independently predictive of good long-term outcome. CONCLUSIONS This research is the first to study endogenous changes in plasma α-MSH after stroke. The independent effect of early plasma α-MSH on stroke outcome, as well as a growing body of experimental data demonstrating improved stroke outcome with exogenous α-MSH administration, suggests a potential therapeutic role for α-MSH in the treatment of stroke.
Collapse
Affiliation(s)
- Dannielle Zierath
- Department of Neurology, University of Washington School of Medicine, Box 359775, Harborview Medical Center, 325 9th Ave, Seattle, WA 98104-2499, USA
| | | | | | | | | |
Collapse
|
50
|
Soos S, Petervari E, Szekely M, Jech-Mihalffy A, Balasko M. Complex catabolic effects of central alpha-MSH infusion in rats of altered nutritional states: differences from leptin. J Mol Neurosci 2010; 43:209-16. [PMID: 20953734 DOI: 10.1007/s12031-010-9462-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 10/05/2010] [Indexed: 01/15/2023]
Abstract
The hypothalamic melanocortin (MC) system is a major catabolic regulator of energy balance: it suppresses food intake (FI), elevates metabolic rate, and reduces body weight (BW). The primary activator of the MC system [mainly via the alpha-melanocyte stimulating hormone (alpha-MSH)] is the adipocyte-derived leptin. With increasing BW, resistance develops to leptin-induced anorexia, but independent of this, in genetically modified animals, some alpha-MSH actions were maintained. We investigated the responsiveness of the MC system in its complexity (FI vs. metabolic correlates) in genetically intact male Wistar rats of different nutritional states (and different leptin sensitivities), i.e., in rats aged 2 months [normally fed (NF2)] or 6 months [calorie-restricted (CR6), fed ad libitum (NF6), and high-fat diet-induced obese (HF6) groups]. A 7-day-long, 1-μg/μl/h intracerebroventricular infusion of alpha-MSH reduced BW in all groups, particularly in NF6 and NF2 animals, and even CR6 rats lost BW upon alpha-MSH infusion (in contrast to leptin administration). Anorexia developed in NF2-NF6 and less in CR6 groups, and some FI fall was also seen in HF6 rats. The hypermetabolic effects (temperature/heart rate elevations) were most pronounced in CR6 and next in HF6 rats. These data suggest that alpha-MSH responsiveness is maintained in various forms (depending on nutritional state), despite obesity-induced leptin resistance.
Collapse
Affiliation(s)
- Szilvia Soos
- Department of Pathophysiology and Gerontology, Medical School, University of Pecs, 12 Szigeti ut, Pecs H-7624, Hungary
| | | | | | | | | |
Collapse
|