1
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. SCIENCE ADVANCES 2024; 10:eadp3284. [PMID: 39661675 PMCID: PMC11633741 DOI: 10.1126/sciadv.adp3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Light is an important environmental factor for vision and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6 to 8 weeks in male, but not female, mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are substantially attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and better understanding of metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
2
|
Nagao K, Yoshikawa S, Urakami H, Fujita Y, Komura A, Nakashima M, Oh-Hora M, Fujimura A, Hiyama TY, Naruse K, Morizane S, Tominaga M, Takamori K, Miyake S. Ligand-independent function of β2-adrenergic receptor affects IgE-mediated Ca 2+ influx in mast cells. Biochem Biophys Res Commun 2024; 733:150595. [PMID: 39191189 DOI: 10.1016/j.bbrc.2024.150595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Mast cells are key effector cells that elicit immunoglobulin E (IgE)-mediated allergic inflammations. Allergen cross-linking of IgE bound to the high-affinity IgE receptor, FcεRI, on mast cells triggers signaling cascades that activate signal proteins and evoke extracellular Ca2+ influx, which are crucial for cytokine production. The β2-adrenergic receptor (Adrb2) on mast cells negatively regulates FcεRI signaling, as demonstrated by the inhibition of IgE/antigen (Ag)-induced activation by Adrb2 agonists. OBJECTIVE Although β2-adrenergic-related reagents are known to influence mast cell functions, the specific intrinsic role of Adrb2 in these cells is not fully understood, potentially because of off-target effects. In this study, the additional roles of Adrb2 in mast cells were investigated, specifically the involvement of Adrb2 in FcεRI signaling, using Adrb2-/- mice. METHODS Adrb2-/- mice were used to investigate the roles of Adrb2 in mast cells by examining bone marrow-derived mast cells (BMMCs) for surface expression of mast cell markers, granule numbers, and gene expression of mast cell proteases. Cytokine production, Ca2+ influx, and nuclear factor of activated T cells (NFAT) nuclear translocation were measured in Adrb2-/- and Adrb2+/+ BMMCs upon IgE/Ag stimulation. RESULTS Adrb2-/- did not affect the generation of BMMCs, their surface expression of mast cell markers, granule numbers, or gene expression of mast cell proteases, indicating that the absence of Adrb2 had no adverse effect on mast cell development. However, Adrb2-/- BMMCs exhibited reduced tumor necrosis factor α (TNFα) production and diminished Ca2⁺ influx upon IgE/Ag stimulation, which correlated with decreased NFAT translocation. Restoration of Adrb2 in Adrb2-/- BMMCs rescued cytokine production. Notably, FcεRI-mediated phosphorylation of the phospholipase PLCγ1 and mitogen-activated protein kinases (MAPKs) remained unchanged in the absence of Adrb2. CONCLUSION These results suggest that Adrb2 has a novel ligand-independent function, increasing Ca2+ entry in mast cells when stimulated with IgE/Ag.
Collapse
Affiliation(s)
- Kei Nagao
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan; Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Soichiro Yoshikawa
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan; Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.
| | - Hitoshi Urakami
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan; Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yuki Fujita
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ayaka Komura
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Miho Nakashima
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masatsugu Oh-Hora
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Fujimura
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Y Hiyama
- Department of Integrative Physiology, Tottori University Graduate School, And Faculty of Medicine, Yonago, Japan
| | - Keiji Naruse
- Department of Cellular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan; Department of Cardiovascular Physiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsutoshi Tominaga
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Kenji Takamori
- Juntendo Itch Research Center (JIRC), Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan.
| | - Sachiko Miyake
- Department of Immunology, School of Medicine, Juntendo University, Tokyo, Japan.
| |
Collapse
|
3
|
González-Contreras C, Martorell M, Ulloa N, Ochoa-Rosales C, Díaz-Toro F, Petermann-Rocha F, Celis-Morales C, Villagran M, Mardones L. Association between the rs12654778 SNP of the β-2 adrenergic receptor and LDL cholesterol levels in the Chilean adult population. ENDOCRINOL DIAB NUTR 2024; 71:397-404. [PMID: 39608965 DOI: 10.1016/j.endien.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 06/27/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION Various polymorphisms in the beta-2 adrenergic receptor (ADRB2) gene have been associated with cardiometabolic risk factors, such as hypertension, dyslipidemia, type 2 diabetes mellitus and obesity contributing to the physiopathology of these chronic conditions. However, the association of the single nucleotide polymorphism (SNP) rs12654778 at the ADRB2 gene with metabolic changes has been poorly studied and there is no information on the Chilean adult population. OBJECTIVE To investigate the association between the rs12654778 SNP at the ADRB2 gene with cardiometabolic risk markers in a Chilean adult population. METHODS We conducted a cross-sectional study including 404 participants from the GENADIO study whom were genotyped for rs12654778 and categorized into GG, AG, and AA genotypes. Associations with cardiometabolic risk markers, such as blood pressure, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, blood glucose and insulin were examined using multivariate regression analysis, while statistical models were adjusted for sociodemographic and lifestyle variables. RESULTS Our findings indicate a significant association between the presence of the protective genotype (AA) of the rs12654778 polymorphism and lower low-density lipoprotein cholesterol levels corresponding to 8.75mg/dL per each copy of the protective allele (maximally adjusted model). No significant associations were seen for the remaining variables. CONCLUSION The AA genotype of the rs12654778 SNP at the ADRB2 gene had a protective effect specifically against low-density lipoprotein cholesterol levels. This is the first study ever conducted in Chile on this SNP of ADRB2 and one of the few conducted worldwide to establish an association between the rs12654778 SNP at the ADRB2 gene and LDL cholesterol.
Collapse
Affiliation(s)
- Carlos González-Contreras
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile; Centre for Healthy Living, Universidad de Concepción, Chile
| | - Natalia Ulloa
- Centre for Healthy Living, Universidad de Concepción, Chile; Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
| | - Carolina Ochoa-Rosales
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibañez, Santiago de Chile, Chile; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Fanny Petermann-Rocha
- Centro de Investigación Biomédica, Facultad de Medicine, Universidad Diego Portales, Santiago, Chile
| | - Carlos Celis-Morales
- Human Performance Lab, Education, Physical Activity and Health Research Unit, Universidad Católica del Maule, Talca, Chile; Biomedical Sciences Research Laboratory, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Marcelo Villagran
- Centre of Biodiversity and Sustainable Environments (CIBAS), Universidad Católica de la Santísima Concepción, Concepción, Chile.
| | - Lorena Mardones
- Centre of Biodiversity and Sustainable Environments (CIBAS), Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
4
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617839. [PMID: 39416062 PMCID: PMC11483057 DOI: 10.1101/2024.10.11.617839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Light is an important environmental factor for vision, and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6-8 weeks, in male, but not, female mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are significantly attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and inform new approaches for understanding metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| |
Collapse
|
5
|
Lymperopoulos A, Borges JI, Stoicovy RA. Cyclic Adenosine Monophosphate in Cardiac and Sympathoadrenal GLP-1 Receptor Signaling: Focus on Anti-Inflammatory Effects. Pharmaceutics 2024; 16:693. [PMID: 38931817 PMCID: PMC11206770 DOI: 10.3390/pharmaceutics16060693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a multifunctional incretin hormone with various physiological effects beyond its well-characterized effect of stimulating glucose-dependent insulin secretion in the pancreas. An emerging role for GLP-1 and its receptor, GLP-1R, in brain neuroprotection and in the suppression of inflammation, has been documented in recent years. GLP-1R is a G protein-coupled receptor (GPCR) that couples to Gs proteins that stimulate the production of the second messenger cyclic 3',5'-adenosine monophosphate (cAMP). cAMP, acting through its two main effectors, protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac), exerts several anti-inflammatory (and some pro-inflammatory) effects in cells, depending on the cell type. The present review discusses the cAMP-dependent molecular signaling pathways elicited by the GLP-1R in cardiomyocytes, cardiac fibroblasts, central neurons, and even in adrenal chromaffin cells, with a particular focus on those that lead to anti-inflammatory effects by the GLP-1R. Fully elucidating the role cAMP plays in GLP-1R's anti-inflammatory properties can lead to new and more precise targets for drug development and/or provide the foundation for novel therapeutic combinations of the GLP-1R agonist medications currently on the market with other classes of drugs for additive anti-inflammatory effect.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328-2018, USA; (J.I.B.); (R.A.S.)
| | | | | |
Collapse
|
6
|
Kumari R, Pascalau R, Wang H, Bajpayi S, Yurgel M, Quansah K, Hattar S, Tampakakis E, Kuruvilla R. Sympathetic NPY controls glucose homeostasis, cold tolerance, and cardiovascular functions in mice. Cell Rep 2024; 43:113674. [PMID: 38236776 PMCID: PMC10951981 DOI: 10.1016/j.celrep.2024.113674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/17/2023] [Accepted: 01/01/2024] [Indexed: 01/30/2024] Open
Abstract
Neuropeptide Y (NPY) is best known for its effects in the brain as an orexigenic and anxiolytic agent and in reducing energy expenditure. NPY is also co-expressed with norepinephrine (NE) in sympathetic neurons. Although NPY is generally considered to modulate noradrenergic responses, its specific roles in autonomic physiology remain under-appreciated. Here, we show that sympathetic-derived NPY is essential for metabolic and cardiovascular regulation in mice. NPY and NE are co-expressed in 90% of prevertebral sympathetic neurons and only 43% of paravertebral neurons. NPY-expressing neurons primarily innervate blood vessels in peripheral organs. Sympathetic-specific NPY deletion elicits pronounced metabolic and cardiovascular defects in mice, including reductions in insulin secretion, glucose tolerance, cold tolerance, and pupil size and elevated heart rate, while notably, however, basal blood pressure was unchanged. These findings provide insight into target tissue-specific functions of NPY derived from sympathetic neurons and imply its potential involvement in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Raniki Kumari
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Raluca Pascalau
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hui Wang
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheetal Bajpayi
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Maria Yurgel
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kwaku Quansah
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA; Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emmanouil Tampakakis
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
7
|
Varney MJ, Benovic JL. The Role of G Protein-Coupled Receptors and Receptor Kinases in Pancreatic β-Cell Function and Diabetes. Pharmacol Rev 2024; 76:267-299. [PMID: 38351071 PMCID: PMC10877731 DOI: 10.1124/pharmrev.123.001015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024] Open
Abstract
Type 2 diabetes (T2D) mellitus has emerged as a major global health concern that has accelerated in recent years due to poor diet and lifestyle. Afflicted individuals have high blood glucose levels that stem from the inability of the pancreas to make enough insulin to meet demand. Although medication can help to maintain normal blood glucose levels in individuals with chronic disease, many of these medicines are outdated, have severe side effects, and often become less efficacious over time, necessitating the need for insulin therapy. G protein-coupled receptors (GPCRs) regulate many physiologic processes, including blood glucose levels. In pancreatic β cells, GPCRs regulate β-cell growth, apoptosis, and insulin secretion, which are all critical in maintaining sufficient β-cell mass and insulin output to ensure euglycemia. In recent years, new insights into the signaling of incretin receptors and other GPCRs have underscored the potential of these receptors as desirable targets in the treatment of diabetes. The signaling of these receptors is modulated by GPCR kinases (GRKs) that phosphorylate agonist-activated GPCRs, marking the receptor for arrestin binding and internalization. Interestingly, genome-wide association studies using diabetic patient cohorts link the GRKs and arrestins with T2D. Moreover, recent reports show that GRKs and arrestins expressed in the β cell serve a critical role in the regulation of β-cell function, including β-cell growth and insulin secretion in both GPCR-dependent and -independent pathways. In this review, we describe recent insights into GPCR signaling and the importance of GRK function in modulating β-cell physiology. SIGNIFICANCE STATEMENT: Pancreatic β cells contain a diverse array of G protein-coupled receptors (GPCRs) that have been shown to improve β-cell function and survival, yet only a handful have been successfully targeted in the treatment of diabetes. This review discusses recent advances in our understanding of β-cell GPCR pharmacology and regulation by GPCR kinases while also highlighting the necessity of investigating islet-enriched GPCRs that have largely been unexplored to unveil novel treatment strategies.
Collapse
Affiliation(s)
- Matthew J Varney
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
8
|
Dwaib H, Michel MC. Adrenoceptor Expression and Function in the Endocrine Pancreas. Handb Exp Pharmacol 2024; 285:639-664. [PMID: 38872059 DOI: 10.1007/164_2024_717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The sympathetic nervous system plays an important role in the regulation of endocrine pancreatic function, most importantly insulin release. Among the nine adrenoceptor (AR) subtypes, the α2A-AR appears to be the subtype most abundantly expressed in the human pancreas. While α2- and β-AR have opposing effects, the net response to sympathetic stimulation is inhibition of insulin secretion mediated by α2-AR located in the plasma membrane of pancreatic β cells. This inhibition may be present physiologically as evidenced by increased insulin secretion in healthy and diabetic humans and animals in response to α2-AR antagonists, a finding that was confirmed in all studies. Based on such data and on an association of an α2A-AR polymorphism, that increases receptor expression levels, with an elevated risk for diabetes, increased α2A-AR signaling in the pancreatic β cells has been proposed as a risk factor for the development of type 2 diabetes. Thus, the α2A-AR was proposed as a drug target for the treatment of some forms of type 2 diabetes. Drug research and development programs leveraging this mechanism have reached the clinical stage, but none have resulted in an approved medicine due to a limited efficacy. While β-AR agonists can increase circulating insulin levels in vivo, it remains controversial whether this includes a direct effect on β cells or occurs secondary to general metabolic effects. Therefore, the regulation of endocrine pancreatic function is physiologically interesting but may be of limited therapeutic relevance.
Collapse
Affiliation(s)
- Haneen Dwaib
- Department of Clinical Nutrition and Dietetics, Palestine Ahliya University, Bethlehem, Palestine.
| | - Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
9
|
Remark J, Tong J, Lin MJ, Concepcion A, Mareedu S, Babu GJ, Feske S, Lu CP. Neurotransmitter signaling specifies sweat gland stem cell fate through SLN-mediated intracellular calcium regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557066. [PMID: 37745514 PMCID: PMC10515797 DOI: 10.1101/2023.09.10.557066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Sympathetic nerves co-develop with their target organs and release neurotransmitters to stimulate their functions after maturation. Here, we provide the molecular mechanism that during sweat gland morphogenesis, neurotransmitters released from sympathetic nerves act first to promote sweat duct elongation via norepinephrine and followed by acetylcholine to specify sweat gland stem cell fate, which matches the sequence of neurotransmitter switch. Without neuronal signals during development, the basal cells switch to exhibit suprabasal (luminal) cell features. Sarcolipin (SLN), a key regulator of sarcoendoplasmic reticulum (SR) Ca 2+ -ATPase (SERCA), expression is significantly down-regulated in the sweat gland myoepithelial cells upon denervation. Loss of SLN in sweat gland myoepithelial cells leads to decreased intracellular Ca 2+ over time in response to ACh stimulation, as well as upregulation of luminal cell features. In cell culture experiments, we showed that contrary to the paradigm that elevation of Ca 2+ promote epidermal differentiation, specification of the glandular myoepithelial (basal) cells requires high Ca 2+ while lowering Ca 2+ level promotes luminal (suprabasal) cell fate. Our work highlights that neuronal signals not only act transiently for mature sweat glands to function, but also exert long-term impact on glandular stem cell specification through regulating intracellular Ca 2+ dynamics.
Collapse
|
10
|
Bohuslavova R, Fabriciova V, Smolik O, Lebrón-Mora L, Abaffy P, Benesova S, Zucha D, Valihrach L, Berkova Z, Saudek F, Pavlinkova G. NEUROD1 reinforces endocrine cell fate acquisition in pancreatic development. Nat Commun 2023; 14:5554. [PMID: 37689751 PMCID: PMC10492842 DOI: 10.1038/s41467-023-41306-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
NEUROD1 is a transcription factor that helps maintain a mature phenotype of pancreatic β cells. Disruption of Neurod1 during pancreatic development causes severe neonatal diabetes; however, the exact role of NEUROD1 in the differentiation programs of endocrine cells is unknown. Here, we report a crucial role of the NEUROD1 regulatory network in endocrine lineage commitment and differentiation. Mechanistically, transcriptome and chromatin landscape analyses demonstrate that Neurod1 inactivation triggers a downregulation of endocrine differentiation transcription factors and upregulation of non-endocrine genes within the Neurod1-deficient endocrine cell population, disturbing endocrine identity acquisition. Neurod1 deficiency altered the H3K27me3 histone modification pattern in promoter regions of differentially expressed genes, which resulted in gene regulatory network changes in the differentiation pathway of endocrine cells, compromising endocrine cell potential, differentiation, and functional properties.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Laura Lebrón-Mora
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 25250, Vestec, Czechia
| | - Zuzana Berkova
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Frantisek Saudek
- Diabetes Centre, Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, 14021, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, 25250, Vestec, Czechia.
| |
Collapse
|
11
|
Gibbs RL, Swanson RM, Beard JK, Hicks ZM, Most MS, Beer HN, Grijalva PC, Clement SM, Marks-Nelson ES, Schmidt TB, Petersen JL, Yates DT. Daily injection of the β2 adrenergic agonist clenbuterol improved poor muscle growth and body composition in lambs following heat stress-induced intrauterine growth restriction. Front Physiol 2023; 14:1252508. [PMID: 37745251 PMCID: PMC10516562 DOI: 10.3389/fphys.2023.1252508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Intrauterine growth restriction (IUGR) is associated with reduced β2 adrenergic sensitivity, which contributes to poor postnatal muscle growth. The objective of this study was to determine if stimulating β2 adrenergic activity postnatal would rescue deficits in muscle growth, body composition, and indicators of metabolic homeostasis in IUGR offspring. Methods: Time-mated ewes were housed at 40°C from day 40 to 95 of gestation to produce IUGR lambs. From birth, IUGR lambs received daily IM injections of 0.8 μg/kg clenbuterol HCl (IUGR+CLEN; n = 11) or saline placebo (IUGR; n = 12). Placebo-injected controls (n = 13) were born to pair-fed thermoneutral ewes. Biometrics were assessed weekly and body composition was estimated by ultrasound and bioelectrical impedance analysis (BIA). Lambs were necropsied at 60 days of age. Results: Bodyweights were lighter (p ≤ 0.05) for IUGR and IUGR+CLEN lambs than for controls at birth, day 30, and day 60. Average daily gain was less (p ≤ 0.05) for IUGR lambs than controls and was intermediate for IUGR+CLEN lambs. At day 58, BIA-estimated whole-body fat-free mass and ultrasound-estimated loin eye area were less (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. At necropsy, loin eye area and flexor digitorum superficialis muscles were smaller (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. Longissimus dorsi protein content was less (p ≤ 0.05) and fat-to-protein ratio was greater (p ≤ 0.05) for IUGR but not IUGR+CLEN lambs than for controls. Semitendinosus from IUGR lambs had less (p ≤ 0.05) β2 adrenoreceptor content, fewer (p ≤ 0.05) proliferating myoblasts, tended to have fewer (p = 0.08) differentiated myoblasts, and had smaller (p ≤ 0.05) muscle fibers than controls. Proliferating myoblasts and fiber size were recovered (p ≤ 0.05) in IUGR+CLEN lambs compared to IUGR lambs, but β2 adrenoreceptor content and differentiated myoblasts were not recovered. Semitendinosus lipid droplets were smaller (p ≤ 0.05) in size for IUGR lambs than for controls and were further reduced (p ≤ 0.05) in size for IUGR+CLEN lambs. Conclusion: These findings show that clenbuterol improved IUGR deficits in muscle growth and some metabolic parameters even without recovering the deficit in β2 adrenoreceptor content. We conclude that IUGR muscle remained responsive to β2 adrenergic stimulation postnatal, which may be a strategic target for improving muscle growth and body composition in IUGR-born offspring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Dustin T. Yates
- Stress Physiology Laboratory, Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
12
|
Kumari R, Pascalau R, Wang H, Bajpayi S, Yurgel M, Quansah K, Hattar S, Tampakakis E, Kuruvilla R. Sympathetic NPY controls glucose homeostasis, cold tolerance, and cardiovascular functions in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550381. [PMID: 37546870 PMCID: PMC10402010 DOI: 10.1101/2023.07.24.550381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Neuropeptide Y (NPY) is best known for its effects in the brain as an orexigenic and anxiolytic agent and in reducing energy expenditure. NPY is also co-expressed with Norepinephrine (NE) in sympathetic neurons. Although NPY is generally considered to modulate noradrenergic responses, its specific roles in autonomic physiology remain under-appreciated. Here, we show that sympathetic-derived NPY is essential for metabolic and cardiovascular regulation in mice. NPY and NE are co-expressed in 90% of prevertebral sympathetic neurons and only 43% of paravertebral neurons. NPY-expressing neurons primarily innervate blood vessels in peripheral organs. Sympathetic-specific deletion of NPY elicits pronounced metabolic and cardiovascular defects in mice, including reductions in insulin secretion, glucose tolerance, cold tolerance, pupil size, and an elevation in heart rate, while notably, however, basal blood pressure was unchanged. These findings provide new knowledge about target tissue-specific functions of NPY derived from sympathetic neurons and imply its potential involvement in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Raniki Kumari
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Raluca Pascalau
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Hui Wang
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Sheetal Bajpayi
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, 21205, USA
| | - Maria Yurgel
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Kwaku Quansah
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, 21205, USA
| | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Emmanouil Tampakakis
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, 21205, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| |
Collapse
|
13
|
Sekar P, Ventura EF, Dhanapal ACTA, Cheah ESG, Loganathan A, Quen PL, Appukutty M, Taslim NA, Hardinsyah H, Md Noh MF, Lovegrove JA, Givens I, Vimaleswaran KS. Gene-Diet Interactions on Metabolic Disease-Related Outcomes in Southeast Asian Populations: A Systematic Review. Nutrients 2023; 15:2948. [PMID: 37447274 PMCID: PMC10346616 DOI: 10.3390/nu15132948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Diabetes and obesity are chronic diseases that are a burden to low- and middle-income countries. We conducted this systematic review to understand gene-diet interactions affecting the Southeast Asian population's risk of obesity and diabetes. The literature search was performed on Google Scholar and MEDLINE (PubMed) search engines independently by four reviewers who evaluated the eligibility of articles based on inclusion criteria. Out of 19,031 articles, 20 articles examining gene-diet interactions on obesity and/or diabetes-related traits met the inclusion criteria. Three (Malaysia, Indonesia, and Singapore) out of eleven Association of Southeast Asian Nations (ASEAN) countries have conducted studies on gene-diet interactions on obesity and diabetes. From the 20 selected articles, the most common interactions were observed between macronutrients and genetic risk score (GRS) on metabolic disease-related traits in the Malay, Chinese, and Indian ethnicities. Overall, we identified 29 significant gene-diet interactions in the Southeast Asian population. The results of this systematic review demonstrate ethnic-specific gene-nutrient interactions on metabolic-disease-related traits in the Southeast Asian population. This is the first systematic review to explore gene-diet interactions on obesity and diabetes in the Southeast Asian population and further research using larger sample sizes is required for better understanding and framing nutrigenetic approaches for personalized nutrition.
Collapse
Affiliation(s)
- Padmini Sekar
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK; (P.S.); (E.F.V.); (J.A.L.)
| | - Eduard Flores Ventura
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK; (P.S.); (E.F.V.); (J.A.L.)
| | - Anto Cordelia T. A. Dhanapal
- Centre for Biomedical and Nutrition Research, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, Kampar 31900, Malaysia; (A.C.T.A.D.); (E.S.G.C.); (A.L.); (P.L.Q.)
| | - Eddy Seong Guan Cheah
- Centre for Biomedical and Nutrition Research, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, Kampar 31900, Malaysia; (A.C.T.A.D.); (E.S.G.C.); (A.L.); (P.L.Q.)
| | - Annaletchumy Loganathan
- Centre for Biomedical and Nutrition Research, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, Kampar 31900, Malaysia; (A.C.T.A.D.); (E.S.G.C.); (A.L.); (P.L.Q.)
| | - Phoon Lee Quen
- Centre for Biomedical and Nutrition Research, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, Kampar 31900, Malaysia; (A.C.T.A.D.); (E.S.G.C.); (A.L.); (P.L.Q.)
| | - Mahenderan Appukutty
- Faculty of Sports Science and Recreation, Universiti Teknologi MARA, Shah Alam 40450, Malaysia;
- Nutrition Society of Malaysia, Jalan PJS 1/48 off Jalan Klang Lama, Petaling Jaya 46150, Malaysia
| | - Nurpudji Astuti Taslim
- Clinical Nutrition, Faculty of Medicine, Hasanuddin University, Makassar 90245, Indonesia;
| | - Hardinsyah Hardinsyah
- Department of Community Nutrition, Faculty of Human Ecology, IPB University, Bogor 16680, Indonesia;
| | - Mohd Fairulnizal Md Noh
- Institute for Medical Research, National Institutes of Health, Jalan Setia Murni U13/52, Seksyen U13 Setia Alam, Shah Alam 40170, Malaysia;
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK; (P.S.); (E.F.V.); (J.A.L.)
| | - Ian Givens
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, UK;
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK; (P.S.); (E.F.V.); (J.A.L.)
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, UK;
| |
Collapse
|
14
|
Mizuno Y, Nakanishi Y, Kumanogoh A. Pathophysiological functions of semaphorins in the sympathetic nervous system. Inflamm Regen 2023; 43:30. [PMID: 37291626 DOI: 10.1186/s41232-023-00281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Upon exposure to external stressors, the body senses them and activates the sympathetic nervous system (SNS) to maintain the homeostasis, which is known as the "fight-or-flight" response. Recent studies have revealed that the SNS also plays pivotal roles in regulating immune responses, such as hematopoiesis, leukocyte mobilization, and inflammation. Indeed, overactivation of the SNS causes many inflammatory diseases, including cardiovascular diseases, metabolic disorders, and autoimmune diseases. However, the molecular basis essential for SNS-mediated immune regulation is not completely understood. In this review, we focus on axon guidance cues, semaphorins, which play multifaceted roles in neural and immune systems. We summarize the functions of semaphorins in the crosstalk between the SNS and the immune system, exploring its pathophysiological roles.
Collapse
Affiliation(s)
- Yumiko Mizuno
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
| | - Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
15
|
Ruiz-Otero N, Kuruvilla R. Role of Delta/Notch-like EGF-related receptor in blood glucose homeostasis. Front Endocrinol (Lausanne) 2023; 14:1161085. [PMID: 37223028 PMCID: PMC10200888 DOI: 10.3389/fendo.2023.1161085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/18/2023] [Indexed: 05/25/2023] Open
Abstract
Cell-cell interactions are necessary for optimal endocrine functions in the pancreas. β-cells, characterized by the expression and secretion of the hormone insulin, are a major constituent of functional micro-organs in the pancreas known as islets of Langerhans. Cell-cell contacts between β-cells are required to regulate insulin production and glucose-stimulated insulin secretion, which are key determinants of blood glucose homeostasis. Contact-dependent interactions between β-cells are mediated by gap junctions and cell adhesion molecules such as E-cadherin and N-CAM. Recent genome-wide studies have implicated Delta/Notch-like EGF-related receptor (Dner) as a potential susceptibility locus for Type 2 Diabetes in humans. DNER is a transmembrane protein and a proposed Notch ligand. DNER has been implicated in neuron-glia development and cell-cell interactions. Studies herein demonstrate that DNER is expressed in β-cells with an onset during early postnatal life and sustained throughout adulthood in mice. DNER loss in adult β-cells in mice (β-Dner cKO mice) disrupted islet architecture and decreased the expression of N-CAM and E-cadherin. β-Dner cKO mice also exhibited impaired glucose tolerance, defects in glucose- and KCl-induced insulin secretion, and decreased insulin sensitivity. Together, these studies suggest that DNER plays a crucial role in mediating islet cell-cell interactions and glucose homeostasis.
Collapse
Affiliation(s)
- Nelmari Ruiz-Otero
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
16
|
Chibly AM, Patel VN, Aure MH, Pasquale MC, Martin GE, Ghannam M, Andrade J, Denegre NG, Simpson C, Goldstein DP, Liu FF, Lombaert IMA, Hoffman MP. Neurotrophin signaling is a central mechanism of salivary dysfunction after irradiation that disrupts myoepithelial cells. NPJ Regen Med 2023; 8:17. [PMID: 36966175 PMCID: PMC10039923 DOI: 10.1038/s41536-023-00290-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/27/2023] [Indexed: 03/27/2023] Open
Abstract
The mechanisms that prevent regeneration of irradiated (IR) salivary glands remain elusive. Bulk RNAseq of IR versus non-IR human salivary glands showed that neurotrophin signaling is highly disrupted post-radiation. Neurotrophin receptors (NTRs) were significantly upregulated in myoepithelial cells (MECs) post-IR, and single cell RNAseq revealed that MECs pericytes, and duct cells are the main sources of neurotrophin ligands. Using two ex vivo models, we show that nerve growth factor (NGF) induces expression of MEC genes during development, and upregulation of NTRs in adult MECs is associated with stress-induced plasticity and morphological abnormalities in IR human glands. As MECs are epithelial progenitors after gland damage and are required for proper acinar cell contraction and secretion, we propose that MEC-specific upregulation of NTRs post-IR disrupts MEC differentiation and potentially impedes the ability of the gland to regenerate.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary C Pasquale
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gemma E Martin
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mousa Ghannam
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julianne Andrade
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Noah G Denegre
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Colleen Simpson
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - Fei-Fei Liu
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, ON, M5G 2M9, Canada
| | - Isabelle M A Lombaert
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Biologic and Material Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Abstract
The islets of Langerhans are highly organized structures that have species-specific, three-dimensional tissue architecture. Islet architecture is critical for proper hormone secretion in response to nutritional stimuli. Islet architecture is disrupted in all types of diabetes mellitus and in cadaveric islets for transplantation during isolation, culture, and perfusion, limiting patient outcomes. Moreover, recapitulating native islet architecture remains a key challenge for in vitro generation of islets from stem cells. In this review, we discuss work that has led to the current understanding of determinants of pancreatic islet architecture, and how this architecture is maintained or disrupted during tissue remodeling in response to normal and pathological metabolic changes. We further discuss both empirical and modeling data that highlight the importance of islet architecture for islet function.
Collapse
Affiliation(s)
- Melissa T. Adams
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barak Blum
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
- CONTACT Barak Blum Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705, USA
| |
Collapse
|
18
|
Huang Z, Liu X. Network Pharmacology and Molecular Docking Analysis on Targets and Mechanisms of Berberine in Atypical Antipsychotic-Induced Metabolic Syndrome. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221129106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Berberine (BBR), an isoquinoline alkaloid, possesses multiply pharmacological effects as a potential therapeutic drug for antipsychotic-induced metabolic syndrome (MetS). However, the underlying therapeutic mechanisms have not been fully elucidated. In this study, we aim to investigate the possible mechanisms by identifying the key targets and biological pathways through network pharmacology and molecular docking analysis. A total of 23 overlapping targets in the intersection set among BBR, atypical antipsychotic drugs (AADs), and MetS were determined. PPI network analysis showed that 22 out of the 23 overlapping targets closely interacted with the others. The following pathway enrichment analysis and molecular docking indicated a central role of peroxisome proliferator-activated receptor-γ (PPARG) as the key target of BBR against AAD-induced MetS by acting on the PPAR signaling pathway, lipid and atherosclerosis, and AMP activated protein kinase (AMPK) signaling pathway. In addition, cytochrome P-450 2D6 (CYP2D6) could be considered as another target of BBR in ameliorating antipsychotic-induced metabolic side effects. Collectively, this study investigated the central targets and biological pathways of BBR against AAD-induced MetS from a systematic perspective, and thus brings novel insights into further understanding of the protective effects of BBR.
Collapse
Affiliation(s)
- Zhuowei Huang
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, Hubei Province, China
- Department of Traditional Chinese Integrated Western Medicine, Wuhan Hospital for Psychotherapy, Wuhan, Hubei Province, China
| | - Xiaolan Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, Hubei Province, China
- Department of Traditional Chinese Integrated Western Medicine, Wuhan Hospital for Psychotherapy, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [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: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
20
|
Mapps AA, Boehm E, Beier C, Keenan WT, Langel J, Liu M, Thomsen MB, Hattar S, Zhao H, Tampakakis E, Kuruvilla R. Satellite glia modulate sympathetic neuron survival, activity, and autonomic function. eLife 2022; 11:74295. [PMID: 35997251 PMCID: PMC9433091 DOI: 10.7554/elife.74295] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Satellite glia are the major glial cells in sympathetic ganglia, enveloping neuronal cell bodies. Despite this intimate association, the extent to which sympathetic functions are influenced by satellite glia in vivo remains unclear. Here, we show that satellite glia are critical for metabolism, survival, and activity of sympathetic neurons and modulate autonomic behaviors in mice. Adult ablation of satellite glia results in impaired mTOR signaling, soma atrophy, reduced noradrenergic enzymes, and loss of sympathetic neurons. However, persisting neurons have elevated activity, and satellite glia-ablated mice show increased pupil dilation and heart rate, indicative of enhanced sympathetic tone. Satellite glia-specific deletion of Kir4.1, an inward-rectifying potassium channel, largely recapitulates the cellular defects observed in glia-ablated mice, suggesting that satellite glia act in part via K+-dependent mechanisms. These findings highlight neuron–satellite glia as functional units in regulating sympathetic output, with implications for disorders linked to sympathetic hyper-activity such as cardiovascular disease and hypertension.
Collapse
Affiliation(s)
- Aurelia A Mapps
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Erica Boehm
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Corinne Beier
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - William T Keenan
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Jennifer Langel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Michael Liu
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Michael B Thomsen
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | | | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
21
|
Osipovich AB, Stancill JS, Cartailler JP, Dudek KD, Magnuson MA. Excitotoxicity and Overnutrition Additively Impair Metabolic Function and Identity of Pancreatic β-Cells. Diabetes 2020; 69:1476-1491. [PMID: 32332159 PMCID: PMC7809715 DOI: 10.2337/db19-1145] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/20/2020] [Indexed: 12/14/2022]
Abstract
A sustained increase in intracellular Ca2+ concentration (referred to hereafter as excitotoxicity), brought on by chronic metabolic stress, may contribute to pancreatic β-cell failure. To determine the additive effects of excitotoxicity and overnutrition on β-cell function and gene expression, we analyzed the impact of a high-fat diet (HFD) on Abcc8 knockout mice. Excitotoxicity caused β-cells to be more susceptible to HFD-induced impairment of glucose homeostasis, and these effects were mitigated by verapamil, a Ca2+ channel blocker. Excitotoxicity, overnutrition, and the combination of both stresses caused similar but distinct alterations in the β-cell transcriptome, including additive increases in genes associated with mitochondrial energy metabolism, fatty acid β-oxidation, and mitochondrial biogenesis and their key regulator Ppargc1a Overnutrition worsened excitotoxicity-induced mitochondrial dysfunction, increasing metabolic inflexibility and mitochondrial damage. In addition, excitotoxicity and overnutrition, individually and together, impaired both β-cell function and identity by reducing expression of genes important for insulin secretion, cell polarity, cell junction, cilia, cytoskeleton, vesicular trafficking, and regulation of β-cell epigenetic and transcriptional program. Sex had an impact on all β-cell responses, with male animals exhibiting greater metabolic stress-induced impairments than females. Together, these findings indicate that a sustained increase in intracellular Ca2+, by altering mitochondrial function and impairing β-cell identity, augments overnutrition-induced β-cell failure.
Collapse
Affiliation(s)
- Anna B Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN
| | - Jennifer S Stancill
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | | | - Karrie D Dudek
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN
| |
Collapse
|
22
|
Yahaya TO, Salisu T, Abdulrahman YB, Umar AK. Update on the genetic and epigenetic etiology of gestational diabetes mellitus: a review. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2020. [DOI: 10.1186/s43042-020-00054-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Abstract
Background
Many studies have been conducted on the genetic and epigenetic etiology of gestational diabetes mellitus (GDM) in the last two decades because of the disease’s increasing prevalence and role in global diabetes mellitus (DM) explosion. An update on the genetic and epigenetic etiology of GDM then becomes imperative to better understand and stem the rising incidence of the disease. This review, therefore, articulated GDM candidate genes and their pathophysiology for the awareness of stakeholders.
Main body (genetic and epigenetic etiology, GDM)
The search discovered 83 GDM candidate genes, of which TCF7L2, MTNR1B, CDKAL1, IRS1, and KCNQ1 are the most prevalent. Certain polymorphisms of these genes can modulate beta-cell dysfunction, adiposity, obesity, and insulin resistance through several mechanisms. Environmental triggers such as diets, pollutants, and microbes may also cause epigenetic changes in these genes, resulting in a loss of insulin-boosting and glucose metabolism functions. Early detection and adequate management may resolve the condition after delivery; otherwise, it will progress to maternal type 2 diabetes mellitus (T2DM) and fetal configuration to future obesity and DM. This shows that GDM is a strong risk factor for T2DM and, in rare cases, type 1 diabetes mellitus (T1DM) and maturity-onset diabetes of the young (MODY). This further shows that GDM significantly contributes to the rising incidence and burden of DM worldwide and its prevention may reverse the trend.
Conclusion
Mutations and epigenetic changes in certain genes are strong risk factors for GDM. For affected individuals with such etiologies, medical practitioners should formulate drugs and treatment procedures that target these genes and their pathophysiology.
Collapse
|
23
|
Tao X, Hu Y, Li L, Xu R, Fu J, Tong Q, Fu Q. Genetic deletion of β 2 adrenergic receptors exacerbates hepatocellular lipid accumulation in high-fat diet mice. Biochem Biophys Res Commun 2019; 511:73-78. [PMID: 30770098 DOI: 10.1016/j.bbrc.2019.02.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022]
Abstract
β2 Adrenergic receptors (β2ARs) are G protein-coupled receptors (GPCRs) that are expressed in major insulin target tissues. β2ARs play an important role in the regulation of lipid metabolism during aging; however, little is known about the significance of β2ARs in the pathogenesis of hepatic fat accumulation in high-fat diet (HFD) mice. This study aims to examine the role of β2AR in the development of nonalcoholic fatty liver disease (NAFLD) induced by HFD and the underlying mechanisms. Surprisingly, we found that genetic deletion of β2AR significantly increased the liver weight of mice fed a HFD for 20 weeks compared to that of wild-type (WT) mice. Moreover, genetic deletion of β2AR could aggravate HFD-induced liver lipid accumulation and liver injury in mice. Mechanistically, we demonstrated that β2AR deletion significantly activated PPARγ/CD36 signaling via inactivation of the cAMP response element-binding (CREB) protein to facilitate hepatocellular lipid deposition in HFD mice. Together, our results identify β2AR as a plausible therapeutic target for preventing or treating NAFLD.
Collapse
Affiliation(s)
- Xiang Tao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China; Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yuting Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China; Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China.
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China.
| |
Collapse
|