1
|
Makio T, Chen J, Simmen T. ER stress as a sentinel mechanism for ER Ca 2+ homeostasis. Cell Calcium 2024; 124:102961. [PMID: 39471738 DOI: 10.1016/j.ceca.2024.102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Endoplasmic reticulum (ER) stress is triggered upon the interference with oxidative protein folding that aims to produce fully folded, disulfide-bonded and glycosylated proteins, which are then competent to exit the ER. Many of the enzymes catalyzing this process require the binding of Ca2+ ions, including the chaperones BiP/GRP78, calnexin and calreticulin. The induction of ER stress with a variety of drugs interferes with chaperone Ca2+ binding, increases cytosolic Ca2+through the opening of ER Ca2+ channels, and activates store-operated Ca2+ entry (SOCE). Posttranslational modifications (PTMs) of the ER Ca2+ handling proteins through ER stress-dependent phosphorylation or oxidation control these mechanisms, as demonstrated in the case of the sarco/endoplasmic reticulum ATPase (SERCA), inositol 1,4,5 trisphosphate receptors (IP3Rs) or stromal interaction molecule 1 (STIM1). Their aim is to restore ER Ca2+ homeostasis but also to increase Ca2+ transfer from the ER to mitochondria during ER stress. This latter function boosts ER bioenergetics, but also triggers apoptosis if ER Ca2+ signaling persists. ER Ca2+ toolkit oxidative modifications upon ER stress can occur within the ER lumen or in the adjacent cytosol. Enzymes involved in this redox control include ER oxidoreductin 1 (ERO1) or the thioredoxin-family protein disulfide isomerases (PDI) and ERp57. A tight, but adaptive connection between ER Ca2+ content, ER stress and mitochondrial readouts allows for the proper functioning of many tissues, including skeletal muscle, the liver, and the pancreas, where ER stress either maintains or compromises their function, depending on its extent and context. Upon mutation of key regulators of ER Ca2+ signaling, diseases such as muscular defects (e.g., from mutated selenoprotein N, SEPN1/SELENON), or diabetes (e.g., from mutated PERK) are the result.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada.
| |
Collapse
|
2
|
Dahl R, Bezprozvanny I. SERCA pump as a novel therapeutic target for treating neurodegenerative disorders. Biochem Biophys Res Commun 2024; 734:150748. [PMID: 39340928 DOI: 10.1016/j.bbrc.2024.150748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
The neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and Spinocerebellar ataxias (SCAs), present an enormous medical, social, financial and scientific problem. Despite intense research into the causes of these disorders, only marginal progress has been made in the clinic and no cures exist for any of them. Most of the scientific effort has been focused on identification of the major causes of these diseases and on developing ways to target them, such as targeting amyloid accumulation for AD or targeting expression of mutant Huntingtin for HD. Calcium (Ca2+) signaling has long been proposed to play an important role in the pathogenesis of neurodegenerative disorders, but blockers of Ca2+ channels and Ca2+ signaling proteins have not been translated to clinic primarily due to side effects related to the important roles of target molecules for these compounds at the peripheral tissues. In this review article, we would like to discuss an idea that recently identified positive allosteric modulators (PAMs) of the sarco-endoplasmic reticulum calcium (SERCA) pump may provide a promising approach to develop therapeutic compounds for treatment of these disorders. This hypothesis is supported by the preclinical data obtained with animal models of AD and PD. The first critical test of this idea will be an imminent phase I study that will offer an opportunity to evaluate potential side effects of this class of compounds in humans.
Collapse
Affiliation(s)
- Russell Dahl
- Neurodon Corporation, 9800 Connecticut Drive, Crown Point, IN, 46307, USA.
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnical University, St. Petersburg, Russia.
| |
Collapse
|
3
|
Uddin MN, Thomas DW. SERCA Modulators Reveal Distinct Signaling and Functional Roles of T Lymphocyte Ca 2+ Stores. Int J Mol Sci 2024; 25:12095. [PMID: 39596161 PMCID: PMC11593871 DOI: 10.3390/ijms252212095] [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: 10/19/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
The allosteric SERCA (Sarcoplasmic/Endoplasmic Reticulum Ca2+-ATPase) activator CDN1163 has been recently added to the group of pharmacological tools for probing SERCA function. We chose to investigate the effects of the compound on T lymphocyte Ca2+ stores, using the well-described Jurkat T lymphocyte as a reliable cell system for Ca2+ signaling pathways. Our study identified the lowest concentrations of the SERCA inhibitors thapsigargin (TG) and 2,5-di-(tert butyl)-1,4-benzohydroquinone (tBHQ) capable of releasing Ca2+, permitting the differentiation of the TG-sensitive SERCA 2b Ca2+ store from the tBHQ-sensitive SERCA 3 Ca2+ store. We proceeded to test the effects of CDN1163 on Ca2+ stores, examining specific actions on the SERCA 2b and SERCA 3 Ca2+ pools using our low-dose SERCA blocker regimen. In contrast to previous work, we find CDN1163 exerts complex time-sensitive and SERCA isoform-specific actions on Ca2+ stores. Surprisingly, short-term exposure (0-30 min) to CDN1163 perturbs T cell Ca2+ stores by suppressing Ca2+ uptake with diminished Ca2+ release from the SERCA 2b-controlled store. Concomitantly, we find evidence for a SERCA-activating effect of CDN1163 on the SERCA-3 regulated store, given the observation of increased Ca2+ release inducible by low-dose tBHQ. Intriguingly, longer-term (>12 h) CDN1163 exposure reversed this pattern, with increased Ca2+ release from SERCA 2b-regulated pools yet decreased Ca2+ release responses from the tBHQ-sensitive SERCA 3 pool. Indeed, this remodeling of SERCA 2b Ca2+ stores with longer-term CDN1163 exposure also translated into the compound's ability to protect Jurkat T lymphocytes from TG but not tBHQ-induced growth suppression.
Collapse
Affiliation(s)
| | - David W. Thomas
- Department of Pharmaceutical Sciences, Thomas J. Long School of Pharmacy University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
4
|
Heger V, Benesova B, Majekova M, Rezbarikova P, Hunyadi A, Horakova L, Viskupicova J. Polyphenolic Compounds Activate SERCA1a and Attenuate Methylglyoxal- and Palmitate-Induced Impairment in Pancreatic INS-1E Beta Cells. Cells 2024; 13:1860. [PMID: 39594609 PMCID: PMC11593225 DOI: 10.3390/cells13221860] [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: 10/07/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) is an important regulatory protein responsible for maintaining calcium homeostasis within cells. Impairment of SERCA associated with activity/expression decrease has been implicated in multiple chronic conditions, including cardiovascular diseases, diabetes, cancer, neurodegenerative diseases, and skeletal muscle pathologies. Natural polyphenols have been recognized to interact with several target proteins involving SERCA. To date, only a limited number of polyphenolic compounds or their derivatives have been described either to increase SERCA activity/expression directly or to affect Ca2+ signaling pathways. In this study, we tested polyphenols for their ability to activate SERCA1a in the absence or presence of methylglyoxal or palmitate and to impact insulin release in pancreatic beta cells. The protective effects of these compounds against methylglyoxal- or palmitate-induced injury were evaluated. Results indicate that 6-gingerol, resveratrol, and ellagic acid activate SERCA1a and protect against activity decrease induced by methylglyoxal and palmitate. Molecular docking analysis revealed the binding of these polyphenols to Glu439 in the SERCA1a P-domain, suggesting a critical role in the stimulation of enzyme activity. Ellagic acid was found to directly stimulate the activity of SERCA1a, marking the first instance of such an observation.
Collapse
Affiliation(s)
- Vladimir Heger
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Barbora Benesova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
- Faculty of Natural Sciences, Comenius University, 841 04 Bratislava, Slovakia
| | - Magdalena Majekova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Petronela Rezbarikova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, H-6720 Szeged, Hungary;
| | - Lubica Horakova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| | - Jana Viskupicova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine of the Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (V.H.); (B.B.); (M.M.); (P.R.); (L.H.)
| |
Collapse
|
5
|
Sirizi MAG, Esmailidehaj M, Mohamadi-Zarch SM, Yadeghari M, Azizian H. Cardioprotective effects of GPER agonist in ovariectomized diabetic rats: reversing ER stress and structural changes. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03438-4. [PMID: 39298018 DOI: 10.1007/s00210-024-03438-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 09/06/2024] [Indexed: 09/21/2024]
Abstract
The incidence of diabetic cardiomyopathy (DCM) significantly increases in postmenopausal women, suggesting protective roles of estrogen. Excessive endoplasmic reticulum (ER) stress alters myocardial structure, which plays a crucial role in DCM. The G protein-coupled estrogen receptor (GPER) has been demonstrated to have cardioprotective effects, but it remains unclear whether these effects involve the amelioration of structural changes induced by ER stress. The objective of this study was to determine whether GPER can prevent cardiac structural changes by attenuating ER stress. Female ovariectomized (OVX) rats were divided into three groups: OVX, OVX + T2D, and OVX + T2D + G1. T2D was induced by a high-fat diet, and streptozotocin and G1, a GPER agonist, were administered for 6 weeks. Finally, histological changes of the myocardium were examined and the expression of sarcoplasmic reticulum calcium ATPase (SERCA2α), GRP78 as an ER stress marker, and apoptotic signalings were determined by Western blot. We observed that the induction of T2D resulted in an increased cardiac weight index, left ventricular wall thickness, and myocyte diameter. However, GPER activation reversed these changes. T2D increased cardiac protein levels of GRP78, caspase-12, and Bax, while decreasing levels of SERCA2α and Bcl-2. Nevertheless, GPER activation reduced the expression of GRP78 in OVX + T2D rats. Furthermore, GPER activation significantly reduced cardiac caspase-12 and Bax levels and increased SERCA2α and Bcl-2 expression. In conclusion, our data suggest that GPER activation ameliorates DCM by inhibiting ER stress-induced cardiac structural changes. These findings provide a new potential target for therapeutic intervention and drug discovery specifically tailored for postmenopausal diabetic women.
Collapse
Affiliation(s)
- Mohammad Amin Ghaffari Sirizi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mansour Esmailidehaj
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyed-Mahdi Mohamadi-Zarch
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Maryam Yadeghari
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
- Department of Anatomy and Cell BioloAAgy, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Azizian
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
6
|
Harmon RM, Ayers JL, McCarthy EF, Kowalczyk AP, Green KJ, Simpson CL. Pumping the Breaks on Acantholytic Skin Disorders: Targeting Calcium Pumps, Desmosomes, and Downstream Signaling in Darier, Hailey-Hailey, and Grover Disease. J Invest Dermatol 2024:S0022-202X(24)01925-0. [PMID: 39207315 DOI: 10.1016/j.jid.2024.06.1289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 09/04/2024]
Abstract
Acantholytic skin disorders, by definition, compromise intercellular adhesion between epidermal keratinocytes. The root cause of blistering in these diseases traces back to direct disruption of adhesive cell-cell junctions, exemplified by autoantibody-mediated attack on desmosomes in pemphigus. However, genetic acantholytic disorders originate from more indirect mechanisms. Darier disease and Hailey-Hailey disease arise from mutations in the endoplasmic reticulum calcium pump, SERCA2, and the Golgi calcium/manganese pump, SPCA1, respectively. Though the disease-causing mutations have been known for nearly 25 years, the mechanistic linkage between dysregulation of intracellular ion stores and weakening of cell-cell junctions at the plasma membrane remains puzzling. The molecular underpinnings of a related idiopathic disorder, Grover disease, are even less understood. Due to an incomplete understanding of acantholytic pathology at the molecular level, these disorders lack proven, targeted treatment options, leaving patients with the significant physical and psychological burdens of chronic skin blistering, infections, and pain. This article aims to review what is known at the molecular, cellular, and clinical levels regarding these under-studied disorders and to highlight knowledge gaps and promising ongoing research. Armed with this knowledge, our goal is to aid investigators in defining essential questions about disease pathogenesis and to accelerate progress toward novel therapeutic strategies.
Collapse
Affiliation(s)
- Robert M Harmon
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Jessica L Ayers
- Molecular Medicine and Mechanisms of Disease PhD Program, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA; Department of Dermatology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Erin F McCarthy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Andrew P Kowalczyk
- Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cory L Simpson
- Department of Dermatology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
7
|
Liiv M, Vaarmann A, Safiulina D, Choubey V, Gupta R, Kuum M, Janickova L, Hodurova Z, Cagalinec M, Zeb A, Hickey MA, Huang YL, Gogichaishvili N, Mandel M, Plaas M, Vasar E, Loncke J, Vervliet T, Tsai TF, Bultynck G, Veksler V, Kaasik A. ER calcium depletion as a key driver for impaired ER-to-mitochondria calcium transfer and mitochondrial dysfunction in Wolfram syndrome. Nat Commun 2024; 15:6143. [PMID: 39034309 PMCID: PMC11271478 DOI: 10.1038/s41467-024-50502-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 07/10/2024] [Indexed: 07/23/2024] Open
Abstract
Wolfram syndrome is a rare genetic disease caused by mutations in the WFS1 or CISD2 gene. A primary defect in Wolfram syndrome involves poor ER Ca2+ handling, but how this disturbance leads to the disease is not known. The current study, performed in primary neurons, the most affected and disease-relevant cells, involving both Wolfram syndrome genes, explains how the disturbed ER Ca2+ handling compromises mitochondrial function and affects neuronal health. Loss of ER Ca2+ content and impaired ER-mitochondrial contact sites in the WFS1- or CISD2-deficient neurons is associated with lower IP3R-mediated Ca2+ transfer from ER to mitochondria and decreased mitochondrial Ca2+ uptake. In turn, reduced mitochondrial Ca2+ content inhibits mitochondrial ATP production leading to an increased NADH/NAD+ ratio. The resulting bioenergetic deficit and reductive stress compromise the health of the neurons. Our work also identifies pharmacological targets and compounds that restore Ca2+ homeostasis, enhance mitochondrial function and improve neuronal health.
Collapse
Affiliation(s)
- Mailis Liiv
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Annika Vaarmann
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.
| | - Dzhamilja Safiulina
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Vinay Choubey
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Ruby Gupta
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Malle Kuum
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Lucia Janickova
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Ch. du Musée 14, 1700, Fribourg, Switzerland
- Department of Cell Pharmacology and Developmental Toxicology, Institute of Experimental Pharmacology and Toxicology, Dúbravská cesta 9, 84104, Bratislava, Slovakia
| | - Zuzana Hodurova
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
- Department of Cell Pharmacology and Developmental Toxicology, Institute of Experimental Pharmacology and Toxicology, Dúbravská cesta 9, 84104, Bratislava, Slovakia
| | - Michal Cagalinec
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
- Department of Cellular Cardiology, Institute of Experimental Endocrinology, Biomedical Research Center and Centre of Excellence for Advanced Materials Application, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Akbar Zeb
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Miriam A Hickey
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Yi-Long Huang
- Department of Life Sciences, Institute of Genome Sciences and Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong St., Section 2, Peitou, Taipei, 11221, Taiwan
| | - Nana Gogichaishvili
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Merle Mandel
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Mario Plaas
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Eero Vasar
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia
| | - Jens Loncke
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| | - Ting-Fen Tsai
- Department of Life Sciences, Institute of Genome Sciences and Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong St., Section 2, Peitou, Taipei, 11221, Taiwan
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, O&N1 Herestraat 49, Leuven, Belgium
| | - Vladimir Veksler
- Laboratory of Signaling and Cardiovascular Pathophysiology, Université Paris-Saclay, Inserm, UMR-S 1180, 91400, Orsay, France
| | - Allen Kaasik
- Departments of Pharmacology and Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 50411, Tartu, Estonia.
| |
Collapse
|
8
|
Li H, Pan W, Li C, Cai M, Shi W, Ren Z, Lu H, Zhou Q, Shen H. Heat stress induces calcium dyshomeostasis to subsequent cognitive impairment through ERS-mediated apoptosis via SERCA/PERK/eIF2α pathway. Cell Death Discov 2024; 10:280. [PMID: 38862478 PMCID: PMC11167007 DOI: 10.1038/s41420-024-02047-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Heat exposure is an environmental stressor that has been associated with cognitive impairment. However, the neural mechanisms that underlie this phenomenon have yet to be extensively investigated. The Morris water maze test was utilized to assess cognitive performance. RNA sequencing was employed to discover the primary regulators and pathological pathways involved in cognitive impairment caused by heat. Before heat exposure in vivo and in vitro, activation of the sarco/endoplasmic reticulum (SR/ER) calcium (Ca2+)-ATPase (SERCA) was achieved by CDN1163. Hematoxylin-Eosin, Nissl staining, calcium imaging, transmission electron microscopy, western blot, and immunofluorescence were utilized to visualize histological changes, intracellular calcium levels, endoplasmic reticulum stress (ERS) markers, apoptosis, and synaptic proteins alterations. Heat stress (HS) significantly induced cognitive decline and neuronal damage in mice. By the transcriptome sequencing between control (n = 5) and heat stress (n = 5) mice in hippocampal tissues, we identified a reduction in the expression of the atp2a gene encoding SERCA, accompanied by a corresponding decrease in its protein level. Consequently, this dysregulation resulted in an excessive accumulation of intracellular calcium ions. Furthermore, HS exposure also activated ERS and apoptosis, as evidenced by the upregulation of p-PERK, p-eIF2α, CHOP, and caspase-3. Consistently, a reduction in postsynaptic density protein 95 (PSD95) and synaptophysin (SYN) expressions indicated modifications in synaptic function. Notably, the impacts on neurons caused by HS were found to be mitigated by CDN1163 treatment both in vivo and in vitro. Additionally, SERCA-mediated ERS-induced apoptosis was attenuated by GSK2606414 treatment via inhibiting PERK-eIF2α-CHOP axis that not only curtailed the level of caspase-3 but also elevated the levels of PSD95 and SYN. These findings highlight the significant impact of heat stress on cognitive impairment, and further elucidate the underlying mechanism involving SERCA/PERK/eIF2α pathway.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Wenlan Pan
- Department of Clinical Nutrition, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, China
| | - Chenqi Li
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- Department of Nutrition, The Third Affiliated Hospital of Naval Medical University, Shanghai, 200438, China
| | - Mengyu Cai
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Wenjing Shi
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zifu Ren
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Hongtao Lu
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Qicheng Zhou
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Hui Shen
- Department of Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
9
|
Bednarski TK, Rahim M, Hasenour CM, Banerjee DR, Trenary IA, Wasserman DH, Young JD. Pharmacological SERCA activation limits diet-induced steatohepatitis and restores liver metabolic function in mice. J Lipid Res 2024; 65:100558. [PMID: 38729350 PMCID: PMC11179628 DOI: 10.1016/j.jlr.2024.100558] [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: 01/25/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease is the most common form of liver disease and poses significant health risks to patients who progress to metabolic dysfunction-associated steatohepatitis. Fatty acid overload alters endoplasmic reticulum (ER) calcium stores and induces mitochondrial oxidative stress in hepatocytes, leading to hepatocellular inflammation and apoptosis. Obese mice have impaired liver sarco/ER Ca2+-ATPase (SERCA) function, which normally maintains intracellular calcium homeostasis by transporting Ca2+ ions from the cytoplasm to the ER. We hypothesized that restoration of SERCA activity would improve diet-induced steatohepatitis in mice by limiting ER stress and mitochondrial dysfunction. WT and melanocortin-4 receptor KO (Mc4r-/-) mice were placed on either chow or Western diet (WD) for 8 weeks. Half of the WD-fed mice were administered CDN1163 to activate SERCA, which reduced liver fibrosis and inflammation. SERCA activation also restored glucose tolerance and insulin sensitivity, improved histological markers of metabolic dysfunction-associated steatohepatitis, increased expression of antioxidant enzymes, and decreased expression of oxidative stress and ER stress genes. CDN1163 decreased hepatic citric acid cycle flux and liver pyruvate cycling, enhanced expression of mitochondrial respiratory genes, and shifted hepatocellular [NADH]/[NAD+] and [NADPH]/[NADP+] ratios to a less oxidized state, which was associated with elevated PUFA content of liver lipids. In sum, the data demonstrate that pharmacological SERCA activation limits metabolic dysfunction-associated steatotic liver disease progression and prevents metabolic dysfunction induced by WD feeding in mice.
Collapse
Affiliation(s)
- Tomasz K Bednarski
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mohsin Rahim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Clinton M Hasenour
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Deveena R Banerjee
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
10
|
Dobson JR, Jacobson DA. Disrupted Endoplasmic Reticulum Ca 2+ Handling: A Harβinger of β-Cell Failure. BIOLOGY 2024; 13:379. [PMID: 38927260 PMCID: PMC11200644 DOI: 10.3390/biology13060379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024]
Abstract
The β-cell workload increases in the setting of insulin resistance and reduced β-cell mass, which occurs in type 2 and type 1 diabetes, respectively. The prolonged elevation of insulin production and secretion during the pathogenesis of diabetes results in β-cell ER stress. The depletion of β-cell Ca2+ER during ER stress activates the unfolded protein response, leading to β-cell dysfunction. Ca2+ER is involved in many pathways that are critical to β-cell function, such as protein processing, tuning organelle and cytosolic Ca2+ handling, and modulating lipid homeostasis. Mutations that promote β-cell ER stress and deplete Ca2+ER stores are associated with or cause diabetes (e.g., mutations in ryanodine receptors and insulin). Thus, improving β-cell Ca2+ER handling and reducing ER stress under diabetogenic conditions could preserve β-cell function and delay or prevent the onset of diabetes. This review focuses on how mechanisms that control β-cell Ca2+ER are perturbed during the pathogenesis of diabetes and contribute to β-cell failure.
Collapse
Affiliation(s)
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA;
| |
Collapse
|
11
|
Ali SI, Elkhalifa AME, Nabi SU, Hayyat FS, Nazar M, Taifa S, Rakhshan R, Shah IH, Shaheen M, Wani IA, Muzaffer U, Shah OS, Makhdoomi DM, Ahmed EM, Khalil KAA, Bazie EA, Zawbaee KI, Al Hasan Ali MM, Alanazi RJ, Al Bataj IA, Al Gahtani SM, Salwi AJ, Alrodan LS. Aged garlic extract preserves beta-cell functioning via modulation of nuclear factor kappa-B (NF-κB)/Toll-like receptor (TLR)-4 and sarco endoplasmic reticulum calcium ATPase (SERCA)/Ca 2+ in diabetes mellitus. Diabetol Metab Syndr 2024; 16:110. [PMID: 38778421 PMCID: PMC11110209 DOI: 10.1186/s13098-024-01350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Peripheral insulin resistance and compromised insulin secretion from pancreatic β-cells are significant factors and pathogenic hallmarks of diabetes mellitus (DM). NF-κβ/TLR-4 and SERCA/Ca2+ pathways have been identified as potential pathways regulating insulin synthesis by preserving pancreatic β-cell functioning. The current study aimed to evaluate the therapeutic effect of aged garlic extract (AGE) against DM in a streptozotocin (STZ)-induced rat model with particular emphasis on pancreatic β-cell functioning. METHODS AGE was characterized by gas chromatography-mass spectrometry (GC-MS), Fourier-transform infrared spectroscopy (FTIR), and scanning electron microscopy (SEM) to evaluate its physio-chemical characteristics followed by in-vitro anti-diabetic and antioxidant potential. This was followed by the induction of DM in laboratory animals for investigating the therapeutic action of AGE by evaluating the role of NF-κβ/TLR-4 and the SERCA/Ca2+ pathway. The parameters assessed in the present experimental setup encompassed antioxidant parameters, metabolic indicators, insulin concentration, intracellular calcium levels, apoptotic markers (CCK-8 and Caspase Glo-8), and protein expression (P-62 and APACHE-II). RESULTS AGE characterization by SEM, GC-MS, and X-ray diffraction (XRD) revealed the presence of phenylalanine, alliin, S-allylmercaptocysteine (SAMC), tryptophan, 1-methyl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic acid as major bioactive constituents of AGE. Metabolic studies, including intraperitoneal glucose tolerance test (IPGTT), revealed significantly lower blood glucose levels in the AGE group compared to the disease control group. In contrast, the intraperitoneal insulin tolerance test (ITT) exhibited no significant difference in insulin sensitivity between the AGE supplementation group and the DM control group. Interestingly, AGE was found to have no significant effect on fasting glucose and serum insulin levels. In contrast, AGE supplementation was found to cause significant hypoglycaemia in postprandial blood glucose and insulin levels. Importantly, AGE causes restoration of intracellular Ca2+ levels by modulation of SERCA/Ca2 functioning and inhibition NF-κB/TLR-4 pathway. AGE was found to interact with and inhibit the DR-5/ caspase-8/3 apoptotic complex. Furthermore, microscopic studies revealed degeneration and apoptotic changes in pancreatic β-cells of the DM control group, while supplementation of AGE resulted in inhibition of apoptotic pathway and regeneration of pancreatic β-cells. CONCLUSION The current study suggests that AGE enhance glucose homeostasis by exerting their effects on pancreatic β-cells, without ameliorating peripheral sensitivity. Moreover, AGEs promote an increase in β-cell mass by mitigating the apoptosis of pancreatic β-cells. These findings suggest that AGE could aid in developing a viable alternative therapy for diabetes mellitus (DM).
Collapse
Affiliation(s)
- Sofi Imtiyaz Ali
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Ahmed M E Elkhalifa
- Department of Public Health, College of Health Sciences, Saudi Electronic University, 11673, Riyadh, Saudi Arabia.
- Department of Haematology, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, 1158, Sudan.
| | - Showkat Ul Nabi
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India.
| | | | - Mehak Nazar
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Syed Taifa
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Rabia Rakhshan
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir, 190006, India
| | - Iqra Hussain Shah
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Muzaffer Shaheen
- Preclinical Research Laboratory, Department of Clinical Veterinary Medicine, Ethics and Jurisprudence, Faculty of Veterinary Sciences and Animal Husbandry, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Imtiyaz Ahmad Wani
- Department of Endocrinology and Clinical Research, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, 190002, India
| | - Umar Muzaffer
- Department of Medicine, Govt. Medical College, Srinagar, Jammu and Kashmir, India
| | - Ovais Shabir Shah
- Department of Sheep Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Dil Mohammad Makhdoomi
- Directorate of Extension, Sher-E-Kashmir University of Agricultural Sciences and Technology (SKUAST-Kashmir), Srinagar, Jammu and Kashmir, 190006, India
| | - Elsadig Mohamed Ahmed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Khalil A A Khalil
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Elsharif A Bazie
- Pediatric Department, Faculty of Medicine, University of El Imam El Mahdi, Kosti, 1158, Sudan
| | - Khalid Ibrahim Zawbaee
- Department of Blood Bank, Autonomous University of Barcelona, Al-Ghad International College for Applied Sciences, 155166, Riyadh, Saudi Arabia
| | - Moataz Mohamed Al Hasan Ali
- Department of Pathology, Faculty of Medicine, Al-Baha University, Al-Baha, Saudi Arabia
- Department of Pathology, Faculty of Medicine, University of El Imam El Mahdi, Kosti, 1158, Sudan
| | - Rakan J Alanazi
- Department of Pharmacy Practice, College of Pharmacy, Alfaisal University, 50927, Riyadh, Saudi Arabia
| | | | - Saeed Musfar Al Gahtani
- Department of Blood Bank, College of Applied Medical Sciences, University of King Saud, 11433, Riyadh, Saudi Arabia
| | - Ali Jubran Salwi
- Department of Blood Bank, College of Applied Medical Sciences, University of King Saud, 11433, Riyadh, Saudi Arabia
| | - Lina Saeed Alrodan
- Department of Blood Bank, College of Applied Medical Sciences, University of King Saud, 11433, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Pang SJ, Liu TT, Pan JC, Man QQ, Song S, Zhang J. The Association between the Plasma Phospholipid Profile and Insulin Resistance: A Population-Based Cross-Section Study from the China Adult Chronic Disease and Nutrition Surveillance. Nutrients 2024; 16:1205. [PMID: 38674894 PMCID: PMC11054597 DOI: 10.3390/nu16081205] [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: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The dysfunction of phospholipid metabolism enzymes and the change in membrane phospholipid composition are associated with insulin resistance, indicating that phospholipids play an important role in the regulation of insulin sensitivity. The reflection of phospholipid changes in blood might provide clues for both mechanism understanding and intervention. Using a targeted phospholipidomic approach, 199 phospholipid molecular species were identified and quantified in the plasma of 1053 middle-aged participants from a national investigation. The associations of the phospholipid matrix, clusters, and molecular species with insulin resistance were investigated. A significant association was confirmed between the phospholipid matrix and the homeostatic-model assessment of insulin resistance (HOMA-IR) by a distance-based linear model. Furthermore, three clustered phospholipid modules and 32 phospholipid molecular species were associated with HOMA-IR with the strict control of demographic and lifestyle parameters, family history of diabetes, BMI, WC, and blood lipid parameters. The overall decline in lysophosphatidylcholines (LPCs), the decrease in saturated lysophosphatidylethanolamines (LPEs), the decrease in polyunsaturated/plasmenyl phosphatidylcholines (PCs), and the increase in polyunsaturated phatidylethanolamines (PEs) were the prominent characters of plasma phospholipid perturbation associated with insulin resistance. This suggested that PC- and PE-related metabolic pathways were widely involved in the process of insulin resistance, especially the disorder of LPC acylation to diacyl-PC.
Collapse
Affiliation(s)
- Shao-Jie Pang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
- Key Laboratory of Public Nutrition and Health, National Health Commission of the People’s Republic of China, Beijing 100050, China
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Beijing 100015, China;
| | - Ting-Ting Liu
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Jian-Cun Pan
- Heilongjiang Feihe Dairy Co., Ltd., C-16, 10A Jiuxianqiao Rd., Beijing 100015, China;
| | - Qing-Qing Man
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
| | - Jian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, No. 29 of Nanwei Road, Beijing 100050, China; (S.-J.P.); (T.-T.L.); (Q.-Q.M.)
- Key Laboratory of Public Nutrition and Health, National Health Commission of the People’s Republic of China, Beijing 100050, China
| |
Collapse
|
13
|
Wang KY, Wu SM, Yao ZJ, Zhu YX, Han X. Insufficient TRPM5 Mediates Lipotoxicity-induced Pancreatic β-cell Dysfunction. Curr Med Sci 2024; 44:346-354. [PMID: 38517672 DOI: 10.1007/s11596-023-2795-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 08/28/2023] [Indexed: 03/24/2024]
Abstract
OBJECTIVE While the reduction of transient receptor potential channel subfamily M member 5 (TRPM5) has been reported in islet cells from type 2 diabetic (T2D) mouse models, its role in lipotoxicity-induced pancreatic β-cell dysfunction remains unclear. This study aims to study its role. METHODS Pancreas slices were prepared from mice subjected to a high-fat-diet (HFD) at different time points, and TRPM5 expression in the pancreatic β cells was examined using immunofluorescence staining. Glucose-stimulated insulin secretion (GSIS) defects caused by lipotoxicity were mimicked by saturated fatty acid palmitate (Palm). Primary mouse islets and mouse insulinoma MIN6 cells were treated with Palm, and the TRPM5 expression was detected using qRT-PCR and Western blotting. Palm-induced GSIS defects were measured following siRNA-based Trpm5 knockdown. The detrimental effects of Palm on primary mouse islets were also assessed after overexpressing Trpm5 via an adenovirus-derived Trpm5 (Ad-Trpm5). RESULTS HFD feeding decreased the mRNA levels and protein expression of TRPM5 in mouse pancreatic islets. Palm reduced TRPM5 protein expression in a time- and dose-dependent manner in MIN6 cells. Palm also inhibited TRPM5 expression in primary mouse islets. Knockdown of Trpm5 inhibited insulin secretion upon high glucose stimulation but had little effect on insulin biosynthesis. Overexpression of Trpm5 reversed Palm-induced GSIS defects and the production of functional maturation molecules unique to β cells. CONCLUSION Our findings suggest that lipotoxicity inhibits TRPM5 expression in pancreatic β cells both in vivo and in vitro and, in turn, drives β-cell dysfunction.
Collapse
Affiliation(s)
- Kai-Yuan Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Shi-Mei Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Zheng-Jian Yao
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Yun-Xia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
14
|
Kong L, Zhao Q, Jiang X, Hu J, Jiang Q, Sheng L, Peng X, Wang S, Chen Y, Wan Y, Hou S, Liu X, Ma C, Li Y, Quan L, Chen L, Cui B, Li P. Trimethylamine N-oxide impairs β-cell function and glucose tolerance. Nat Commun 2024; 15:2526. [PMID: 38514666 PMCID: PMC10957989 DOI: 10.1038/s41467-024-46829-0] [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: 01/11/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
β-Cell dysfunction and β-cell loss are hallmarks of type 2 diabetes (T2D). Here, we found that trimethylamine N-oxide (TMAO) at a similar concentration to that found in diabetes could directly decrease glucose-stimulated insulin secretion (GSIS) in MIN6 cells and primary islets from mice or humans. Elevation of TMAO levels impairs GSIS, β-cell proportion, and glucose tolerance in male C57BL/6 J mice. TMAO inhibits calcium transients through NLRP3 inflammasome-related cytokines and induced Serca2 loss, and a Serca2 agonist reversed the effect of TMAO on β-cell function in vitro and in vivo. Additionally, long-term TMAO exposure promotes β-cell ER stress, dedifferentiation, and apoptosis and inhibits β-cell transcriptional identity. Inhibition of TMAO production improves β-cell GSIS, β-cell proportion, and glucose tolerance in both male db/db and choline diet-fed mice. These observations identify a role for TMAO in β-cell dysfunction and maintenance, and inhibition of TMAO could be an approach for the treatment of T2D.
Collapse
Affiliation(s)
- Lijuan Kong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Qijin Zhao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Xiaojing Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Li Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Peng
- College of Future Technology, Institute of Molecular Medicine, National Biomedical Imaging Center, Peking University, 100871, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Shusen Wang
- Tianjin First Central Hospital, Tianjin, China
| | - Yibing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Yanjun Wan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Shaocong Hou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Xingfeng Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Chunxiao Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Yan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Quan
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Liangyi Chen
- College of Future Technology, Institute of Molecular Medicine, National Biomedical Imaging Center, Peking University, 100871, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, China.
| |
Collapse
|
15
|
Zhang R, Zhang Q, Cui Z, Huang B, Ma H. Dimethyl fumarate restores Ca 2+ dyshomeostasis through activation of the SIRT1 signal to treat nonalcoholic fatty liver disease. Life Sci 2024; 341:122505. [PMID: 38364937 DOI: 10.1016/j.lfs.2024.122505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by an excessive lipid accumulation in the liver, with a global prevalence of approximately 25 %. While early-stage steatosis is reversible and can be intervened upon, it has the potential to progress to some serious complications, including cirrhosis and even liver cancer. Dimethyl fumarate (DMF), a derivative of fumaric acid shows promise in intervening in certain diseases. However, the precise effect and underlying mechanism of DMF on hepatic steatosis remain unclear. In this study, we demonstrated that DMF mitigates hepatic steatosis in mice subjected to high-fat/high-cholesterol (HFHC) diets. Meanwhile, our in vivo and in vitro results showed that DMF relieves lipid accumulation, oxidative stress, and endoplasmic reticulum (ER) stress. Mechanically, our findings revealed that the effect of DMF on reducing lipid accumulation is linked to the restoration of Ca2+ homeostasis. Furthermore, we found that activation of the SIRT1 signal by DMF plays an important role in correcting the mishandling of the Ca2+ signal, and knockdown of SIRT1 expression reverses the beneficial role of DMF PA-incubated AML12 cells. In conclusion, our results suggested DMF's amelioration of hepatic steatosis is related to the activation of SIRT1-mediated Ca2+ signaling.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Quanwei Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - ZiYi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - BenZeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
16
|
Wang Y, Duan F, Li J, Li X, Xia L, Zhao W, Wang Z, Song X, Chen J, Wang J, Wang Y, Zhang J, Zhang X, Jiao D. Involvement of nucleus accumbens SERCA2b in methamphetamine-induced conditioned place preference. Addict Biol 2024; 29:e13382. [PMID: 38488467 PMCID: PMC11061847 DOI: 10.1111/adb.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/19/2024] [Accepted: 02/12/2024] [Indexed: 03/19/2024]
Abstract
Methamphetamine (METH) is a highly addictive psycho-stimulant that induces addictive behaviour by stimulating increased dopamine release in the nucleus accumbens (NAc). The sarco/endoplasmic reticulum calcium ion transport ATPases (SERCA or ATP2A) is a calcium ion (Ca2+) pump in the endoplasmic reticulum (ER) membrane. SERCA2b is a SERCA subtype mainly distributed in the central nervous system. This study used conditioned place preference (CPP), a translational drug reward model, to observe the effects of SERCA and SERCA2b on METH-CPP in mice. Result suggested that the activity of SERCA was significantly decreased in NAc after METH-CPP. Intraperitoneal SERCA agonist CDN1163 injection or bilateral CDN1163 microinjection in the NAc inhibited METH-CPP formation. SERCA2b overexpression by the Adeno-associated virus can reduce the DA release of NAc and inhibit METH-CPP formation. Although microinjection of SERCA inhibitor thapsigargin in the bilateral NAc did not significantly aggravate METH-CPP, interference with SERCA2b expression in NAc by adeno-associated virus increased DA release and promoted METH-CPP formation. METH reduced the SERCA ability to transport Ca2+ into the ER in SHSY5Y cells in vitro, which was reversed by CDN1163. This study revealed that METH dysregulates intracellular calcium balance by downregulating SERCA2b function, increasing DA release in NAc and inducing METH-CPP formation. Drugs that target SERCA2b may have the potential to treat METH addiction.
Collapse
Affiliation(s)
- Yujing Wang
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Fan Duan
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Junda Li
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Xiangyu Li
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Lingling Xia
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Wei Zhao
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Ze Wang
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Xun Song
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Juan Chen
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Jingjing Wang
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Yue Wang
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Jing Zhang
- School of Mental HealthBengbu Medical UniversityBengbuChina
| | - Xiaochu Zhang
- School of Mental HealthBengbu Medical UniversityBengbuChina
- CAS Key Laboratory of Brain Function and Disease and School of Life SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Dongliang Jiao
- School of Mental HealthBengbu Medical UniversityBengbuChina
| |
Collapse
|
17
|
Wang Y, Wang M, Su H, Song J, Ren M, Hu P, Liu G, Tong X. SERCA2 dysfunction triggers hypertension by interrupting mitochondrial homeostasis and provoking oxidative stress. Free Radic Biol Med 2024; 212:284-294. [PMID: 38163553 DOI: 10.1016/j.freeradbiomed.2023.12.044] [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: 10/03/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND AND AIM Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) is critical in maintaining Ca2+ homeostasis. The cysteine 674 (C674) is the key redox regulatory cysteine in regulating SERCA2 activity, which is irreversibly oxidized in the renal cortex of hypertensive mice. We have reported that the substitution of C674 by serine causes SERCA2 dysfunction and increases blood pressure by induction of endoplasmic reticulum stress (ERS). This study is to explore whether the dysfunction of SERCA2 causes hypertension by interrupting mitochondrial homeostasis and inducing oxidative stress. METHODS & RESULTS We used heterozygous SERCA2 C674S gene mutation knock-in (SKI) mice, where one copy of C674 was substituted by serine to represent partial C674 oxidation. In renal proximal tubule (RPT) cells, the substitution of C674 by serine decreased mitochondrial Ca2+ content, increased mitochondrial membrane potential, ATP content, and reactive oxygen species (ROS), which could be reversed by ERS inhibitor 4-phenylbutyric acid or SERCA2 agonist CDN1163. In SKI RPT cells, the redox modulator Tempol alleviated oxidative stress, downregulated the protein expression of ERS markers and soluble epoxide hydrolase, upregulated the protein expression of dopamine D1 receptor, and reduced Na+/K+- ATPase activity. In SKI mice, SERCA2 agonists CDN1163 and [6]-Gingerol, or the redox modulator Tempol increased urine output and lowered blood pressure. CONCLUSION The irreversible oxidation of C674 is not only an indicator of increased ROS, but also further inducing oxidative stress to cause hypertension. Activation of SERCA2 or inhibition of oxidative stress is beneficial to alleviate hypertension caused by SERCA2 dysfunction.
Collapse
Affiliation(s)
- Yaping Wang
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Min Wang
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Hang Su
- Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563006, China
| | - Jiarou Song
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Minghua Ren
- Department of Urinary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Pingping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China.
| | - Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China; Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
18
|
Atakan MM, Türkel İ, Özerkliğ B, Koşar ŞN, Taylor DF, Yan X, Bishop DJ. Small peptides: could they have a big role in metabolism and the response to exercise? J Physiol 2024; 602:545-568. [PMID: 38196325 DOI: 10.1113/jp283214] [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: 10/19/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Exercise is a powerful non-pharmacological intervention for the treatment and prevention of numerous chronic diseases. Contracting skeletal muscles provoke widespread perturbations in numerous cells, tissues and organs, which stimulate multiple integrated adaptations that ultimately contribute to the many health benefits associated with regular exercise. Despite much research, the molecular mechanisms driving such changes are not completely resolved. Technological advancements beginning in the early 1960s have opened new avenues to explore the mechanisms responsible for the many beneficial adaptations to exercise. This has led to increased research into the role of small peptides (<100 amino acids) and mitochondrially derived peptides in metabolism and disease, including those coded within small open reading frames (sORFs; coding sequences that encode small peptides). Recently, it has been hypothesized that sORF-encoded mitochondrially derived peptides and other small peptides play significant roles as exercise-sensitive peptides in exercise-induced physiological adaptation. In this review, we highlight the discovery of mitochondrially derived peptides and newly discovered small peptides involved in metabolism, with a specific emphasis on their functions in exercise-induced adaptations and the prevention of metabolic diseases. In light of the few studies available, we also present data on how both single exercise sessions and exercise training affect expression of sORF-encoded mitochondrially derived peptides. Finally, we outline numerous research questions that await investigation regarding the roles of mitochondrially derived peptides in metabolism and prevention of various diseases, in addition to their roles in exercise-induced physiological adaptations, for future studies.
Collapse
Affiliation(s)
- Muhammed M Atakan
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - İbrahim Türkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Berkay Özerkliğ
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Şükran N Koşar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Turkey
| | - Dale F Taylor
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| | - Xu Yan
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
- Sarcopenia Research Program, Australia Institute for Musculoskeletal Sciences (AIMSS), Melbourne, Victoria, Australia
| | - David J Bishop
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Makio T, Simmen T. Not So Rare: Diseases Based on Mutant Proteins Controlling Endoplasmic Reticulum-Mitochondria Contact (MERC) Tethering. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241261228. [PMID: 39070058 PMCID: PMC11273598 DOI: 10.1177/25152564241261228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024]
Abstract
Mitochondria-endoplasmic reticulum contacts (MERCs), also called endoplasmic reticulum (ER)-mitochondria contact sites (ERMCS), are the membrane domains, where these two organelles exchange lipids, Ca2+ ions, and reactive oxygen species. This crosstalk is a major determinant of cell metabolism, since it allows the ER to control mitochondrial oxidative phosphorylation and the Krebs cycle, while conversely, it allows the mitochondria to provide sufficient ATP to control ER proteostasis. MERC metabolic signaling is under the control of tethers and a multitude of regulatory proteins. Many of these proteins have recently been discovered to give rise to rare diseases if their genes are mutated. Surprisingly, these diseases share important hallmarks and cause neurological defects, sometimes paired with, or replaced by skeletal muscle deficiency. Typical symptoms include developmental delay, intellectual disability, facial dysmorphism and ophthalmologic defects. Seizures, epilepsy, deafness, ataxia, or peripheral neuropathy can also occur upon mutation of a MERC protein. Given that most MERC tethers and regulatory proteins have secondary functions, some MERC protein-based diseases do not fit into this categorization. Typically, however, the proteins affected in those diseases have dominant functions unrelated to their roles in MERCs tethering or their regulation. We are discussing avenues to pharmacologically target genetic diseases leading to MERC defects, based on our novel insight that MERC defects lead to common characteristics in rare diseases. These shared characteristics of MERCs disorders raise the hope that they may allow for similar treatment options.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Todo N, Hosogi S, Nakamura S, Noriyama K, Tamiya N, Toda Y, Shigeta M, Takayama K, Ashihara E. Cynaropicrin Increases [Ca 2+] i and Ciliary Beat Frequency in Human Airway Epithelial Cells by Inhibiting SERCA. Biol Pharm Bull 2024; 47:2119-2126. [PMID: 39710380 DOI: 10.1248/bpb.b24-00567] [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: 12/24/2024]
Abstract
Mucociliary clearance (MCC) is a host defense mechanism of the respiratory system. Beating cilia plays a crucial role in the MCC process and ciliary beat frequency (CBF) is activated by several factors including elevations of the intracellular cAMP concentration ([cAMP]i), intracellular Ca2+ concentration ([Ca2+]i), and intracellular pH (pHi). In this study, we investigated whether an artichoke-extracted component cynaropicrin could be a beneficial compound for improving MCC. We found that cynaropicrin increased [cAMP]i using A549 cells bearing Pink Flamindo. Then, we also confirmed that cynaropicrin elevates CBF using airway epithelial ciliated cells (AECCs). We next investigated the effects of cynaropicrin on the alternation of [Ca2+]i, and pHi. Cynaropicrin increased [Ca2+]i, but not pHi. Further experiments also found that cynaropicrin increased [cAMP]i primarily by raising [Ca2+]i. To elucidate the mechanisms of cynaropicrin to increase [Ca2+]i, we investigated the alternation of the effects of cynaropicrin on [Ca2+]i using several compounds. BTP-2 and ruthenium red (RuR) inhibited cynaropicrin-induced [Ca2+]i increase and RuR reduced also [cAMP]i. These results suggest that cynaropicrion increased [Ca2+]i by augmenting the Ca2+ influx and that the increase of [cAMP]i by cynaropicrin was induced by [Ca2+]i elevation. Interestingly, cynaropicrin decreased the Ca2+ concentration in the endoplasmic reticulum following inhibition of sarco-endoplasmic reticulum Ca2+-ATPase (SERCA). SERCA activator CDN1163 abolished this effect. Furthermore, RuR and Ca2+-free conditions suppressed the increase of CBF. In conclusion, cynaropicrin inhibits SERCA, induces store-operated calcium entry, and thereby increases CBF.
Collapse
Affiliation(s)
- Nobuhisa Todo
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
| | - Shigekuni Hosogi
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
| | - Seikou Nakamura
- Laboratory of Pharmacognosy, Kyoto Pharmaceutical University
| | - Kouta Noriyama
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
| | - Nobuyo Tamiya
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
- Pulmonary Medicine, Rakuwakai Otowa Hospital
| | - Yuki Toda
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
| | - Masaki Shigeta
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Eishi Ashihara
- Laboratory of Clinical and Translational Physiology, Kyoto Pharmaceutical University
| |
Collapse
|
21
|
Yu W, Zhang Q, Qiu Y, Chen H, Huang X, Xiao L, Xu G, Li S, Hu P, Tong X. CDN1163 alleviates SERCA2 dysfunction-induced pulmonary vascular remodeling by inhibiting the phenotypic transition of pulmonary artery smooth muscle cells. Clin Exp Hypertens 2023; 45:2272062. [PMID: 37899350 DOI: 10.1080/10641963.2023.2272062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
BACKGROUND AND PURPOSE Substitution of Cys674 (C674) in the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) causes SERCA2 dysfunction which leads to activated inositol requiring enzyme 1 alpha (IRE1α) and spliced X-box binding protein 1 (XBP1s) pathway accelerating cell proliferation of pulmonary artery smooth muscle cells (PASMCs) followed by significant pulmonary vascular remodeling resembling human pulmonary hypertension. Based on this knowledge, we intend to investigate other potential mechanisms involved in SERCA2 dysfunction-induced pulmonary vascular remodeling. EXPERIMENTAL APPROACH Heterozygous SERCA2 C674S knock-in (SKI) mice of which half of cysteine in 674 was substituted by serine to mimic the partial irreversible oxidation of C674 were used. The lungs of SKI mice and their littermate wild-type mice were collected for PASMC culture, protein expression, and pulmonary vascular remodeling analysis. RESULTS SERCA2 dysfunction increased intracellular Ca2+ levels, which activated Ca2+-dependent calcineurin (CaN) and promoted the nuclear translocation and protein expression of the nuclear factor of activated T-lymphocytes 4 (NFAT4) in an IRE1α/XBP1s pathway-independent manner. In SKI PASMCs, the scavenge of intracellular Ca2+ by BAPTA-AM or inhibition of CaN by cyclosporin A can prevent PASMC phenotypic transition. CDN1163, a SERCA2 agonist, suppressed the activation of CaN/NFAT4 and IRE1α/XBP1s pathways, reversed the protein expression of PASMC phenotypic transition markers and cell cycle-related proteins, and inhibited cell proliferation and migration when given to SKI PASMCs. Furthermore, CDN1163 ameliorated pulmonary vascular remodeling in SKI mice. CONCLUSIONS AND IMPLICATIONS SERCA2 dysfunction promotes PASMC phenotypic transition and pulmonary vascular remodeling by multiple mechanisms, which could be improved by SERCA2 agonist CDN1163.
Collapse
Affiliation(s)
- Weimin Yu
- Institute of Health Biological Chemical Medication, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Qian Zhang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yixiang Qiu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Hui Chen
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaoyang Huang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Li Xiao
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- Central Clinical School, Monash University, Melbourne, Australia
| | - Pingping Hu
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
22
|
KLOCKE B, MOORE C, OTT H, PITYCHOUTIS PM. Chronic pharmacological activation of SERCA with CDN1163 affects spatial cognitive flexibility but not attention and impulsivity in mice. Behav Pharmacol 2023; 34:477-487. [PMID: 37917567 PMCID: PMC10624114 DOI: 10.1097/fbp.0000000000000756] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Intracellular calcium (Ca2+) homeostasis is critical for many neural processes, including learning, memory and synaptic plasticity. The sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) is among the key regulators that preserve Ca2+ homeostasis in neurons. SERCAs comprise a set of ubiquitously expressed Ca2+ pumps that primarily function to sequester cytosolic Ca2+ into endoplasmic reticular stores. As SERCA has been implicated in the neurobiology of several neuropsychiatric and neurodegenerative diseases, pharmacological harnessing of its function is critical in understanding SERCA's role in brain physiology and pathophysiology. In the current study, we employed the Morris water maze and 5-choice serial reaction time task (5-CSRTT) to investigate the effects of chronic pharmacological activation of SERCA, using the small allosteric SERCA activator CDN1163, on spatial learning and memory, and executive functioning in naive C57BL/6J mice. Our data show that chronic pharmacological SERCA activation with CDN1163 (20 mg/kg) selectively impairs spatial cognitive flexibility and reversal learning in the Morris water maze while leaving executive functions such as attention and impulsivity intact. Present findings contribute to the growing field of the role of SERCA function in the brain and behavior and expand current knowledge on the use of the small allosteric activator CDN1163 as an investigational tool to study the role of SERCA in regulating neurobehavioral processes and as a potential therapeutic candidate for debilitating brain disorders.
Collapse
Affiliation(s)
- Benjamin KLOCKE
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | - Carter MOORE
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | - Hayden OTT
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | | |
Collapse
|
23
|
Nguyen HT, Noriega Polo C, Wiederkehr A, Wollheim CB, Park KS. CDN1163, an activator of sarco/endoplasmic reticulum Ca 2+ ATPase, up-regulates mitochondrial functions and protects against lipotoxicity in pancreatic β-cells. Br J Pharmacol 2023; 180:2762-2776. [PMID: 37277321 DOI: 10.1111/bph.16160] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND AND PURPOSE High levels of Ca2+ in the endoplasmic reticulum (ER), established by the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), are required for protein folding and cell signalling. Excessive ER Ca2+ release or decreased SERCA activity induces unfolded protein accumulation and ER stress in pancreatic β-cells, leading to defective insulin secretion and diabetes. Here we have investigated the consequences of enhancing ER Ca2+ uptake on β-cell survival and function. EXPERIMENTAL APPROACH The effects of SERCA activator, CDN1163, on Ca2+ homeostasis, protein expression, mitochondrial activities, insulin secretion, and lipotoxicity have been studied in mouse pancreatic β-cells and MIN6 cells. KEY RESULTS CDN1163, increased insulin synthesis and exocytosis from islets. CDN1163 also increased the sensitivity of the cytosolic Ca2+ oscillation response to glucose and potentiated it in dispersed and sorted β-cells. CDN1163 augmented the ER and mitochondrial Ca2+ content, the mitochondrial membrane potential, respiration, and ATP synthesis. CDN1163 up-regulated expression of inositol 1,4,5-trisphosphate receptors and antioxidant enzymes, and mitochondrial biogenesis, including peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α). Overexpression of SERCA2a or 2b replicated the effects of CDN1163, while knockdown of SERCA2 abolished the stimulatory actions of CDN1163. In palmitate-treated β-cells, CDN1163 prevented ER Ca2+ depletion, mitochondrial dysfunction, cytosolic and mitochondrial oxidative stress, defective insulin secretion, and apoptotic cell death. CONCLUSIONS AND IMPLICATIONS Activation of SERCA enhanced mitochondrial bioenergetics and antioxidant capability, suppressing the cytotoxic effects of palmitate. Our results suggest that targeting SERCA could be a novel therapeutic strategy to protect β-cells from lipotoxicity and the development of Type 2 diabetes.
Collapse
Affiliation(s)
- Ha Thu Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, South Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Carlos Noriega Polo
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, South Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | | | - Claes B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, South Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, South Korea
| |
Collapse
|
24
|
Sharlo KA, Lvova ID, Tyganov SA, Zaripova KA, Belova SP, Kostrominova TY, Shenkman BS, Nemirovskaya TL. The Effect of SERCA Activation on Functional Characteristics and Signaling of Rat Soleus Muscle upon 7 Days of Unloading. Biomolecules 2023; 13:1354. [PMID: 37759754 PMCID: PMC10526198 DOI: 10.3390/biom13091354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Skeletal muscle abnormalities and atrophy during unloading are accompanied by the accumulation of excess calcium in the sarcoplasm. We hypothesized that calcium accumulation may occur, among other mechanisms, due to the inhibition of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. Consequently, the use of the SERCA activator will reduce the level of calcium in the sarcoplasm and prevent the negative consequences of muscle unloading. Wistar rats were randomly assigned into one of three groups (eight rats per group): control rats with placebo (C), 7 days of unloading/hindlimb suspension with placebo (7HS), and 7 days of unloading treated with SERCA activator CDN1163 (7HSC). After seven days of unloading the soleus muscle, the 7HS group displayed increased fatigue in the ex vivo test, a significant increase in the level of calcium-dependent CaMK II phosphorylation and the level of tropomyosin oxidation, as well as a decrease in the content of mitochondrial DNA and protein, slow-type myosin mRNA, and the percentage of slow-type muscle fibers. All of these changes were prevented in the 7HSC group. Moreover, treatment with CDN1163 blocked a decrease in the phosphorylation of p70S6k, an increase in eEF2 phosphorylation, and an increase in MuRF-1 mRNA expression. Nevertheless, there were no differences in the degree of fast and slow muscle fiber atrophy between the 7HS and 7HSC groups. Conclusion: SERCA activation during 7 days of unloading prevented an increase in soleus fatigue, the decrease of slow-type myosin, mitochondrial markers, and markers of calcium homeostasis but had no effect on muscle atrophy.
Collapse
Affiliation(s)
- Kristina A. Sharlo
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Irina D. Lvova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Sergey A. Tyganov
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Ksenia A. Zaripova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Svetlana P. Belova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana Y. Kostrominova
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine-Northwest, Gary, IN 46202, USA;
| | - Boris S. Shenkman
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana L. Nemirovskaya
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| |
Collapse
|
25
|
Humbert A, Lefebvre R, Nawrot M, Caussy C, Rieusset J. Calcium signalling in hepatic metabolism: Health and diseases. Cell Calcium 2023; 114:102780. [PMID: 37506596 DOI: 10.1016/j.ceca.2023.102780] [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: 02/28/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
The flexibility between the wide array of hepatic functions relies on calcium (Ca2+) signalling. Indeed, Ca2+ is implicated in the control of many intracellular functions as well as intercellular communication. Thus, hepatocytes adapt their Ca2+ signalling depending on their nutritional and hormonal environment, leading to opposite cellular functions, such as glucose storage or synthesis. Interestingly, hepatic metabolic diseases, such as obesity, type 2 diabetes and non-alcoholic fatty liver diseases, are associated with impaired Ca2+ signalling. Here, we present the hepatocytes' toolkit for Ca2+ signalling, complete with regulation systems and signalling pathways activated by nutrients and hormones. We further discuss the current knowledge on the molecular mechanisms leading to alterations of Ca2+ signalling in hepatic metabolic diseases, and review the literature on the clinical impact of Ca2+-targeting therapeutics.
Collapse
Affiliation(s)
- Alexandre Humbert
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Rémy Lefebvre
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Margaux Nawrot
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Cyrielle Caussy
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France; Département Endocrinologie, Diabète et Nutrition, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Jennifer Rieusset
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France.
| |
Collapse
|
26
|
Huang YP, Huang WW, Tsai KF, Shiao LR, Yang ZH, Tseng SY, Lin YH, Chen CY, Chan P, Leung YM. CDN1163, a SERCA activator, causes intracellular Ca 2+ leak, mitochondrial hyperpolarization and cell cycle arrest in mouse neuronal N2A cells. Neurotoxicology 2023; 98:9-15. [PMID: 37429421 DOI: 10.1016/j.neuro.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/11/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVE Activity or expression of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) is diminished in some disease states such as cardiac failure and diabetes mellitus. A newly developed activator of SERCA, CDN1163, reportedly rescued or alleviated pathological conditions attributed to dysfunctional SERCA. We examined whether CDN1163 could relieve mouse neuronal N2A cell growth inhibition caused by cyclopiazonic acid (CPA, SERCA inhibitor). We also examined how CDN1163 affected cytosolic Ca2+, mitochondrial Ca2+ and mitochondrial membrane potential. METHODS Cell viability was measured by MTT assay and trypan blue exclusion test. Cytosolic Ca2+, mitochondrial Ca2+ and mitochondrial membrane potential were measured using fura 2, Rhod-2 and JC-1, respectively, as fluorescent probes. RESULTS CDN1163 (10 μM) itself suppressed cell proliferation, and did not alleviate CPA's inhibitory effect (and vice versa). Cell cycle was arrested at the G1 phase after CDN1163 treatment. CDN1163 treatment caused a slow yet persistent cytosolic [Ca2+] elevation partly due to Ca2+ release from an internal store other than the CPA-sensitive endoplasmic reticulum (ER). Treatment with CDN1163 for 3 h raised mitochondrial Ca2+ level and such increase was suppressed by MCU-i4 (an inhibitor of mitochondria Ca2+ uniporter, MCU), suggesting Ca2+ entered the mitochondrial matrix through MCU. Treatment of cells with CDN1163 up to 2 days resulted in mitochondrial hyperpolarization. CONCLUSION CDN1163 caused internal Ca2+ leak, cytosolic Ca2+ overload, mitochondrial Ca2+ elevation and hyperpolarization, cell cycle arrest and cell growth inhibition.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Wen-Wei Huang
- Department of Internal Medicine, Gastroenterology and Hepatology Section, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Kun-Feng Tsai
- Department of Internal Medicine, Gastroenterology and Hepatology Section, An Nan Hospital, China Medical University, Tainan, Taiwan; Department of Medical Sciences Industry, Chang Jung Christian University, Tainan, Taiwan
| | - Lian-Ru Shiao
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Zih-He Yang
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Shao-Yun Tseng
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Yu-Hsien Lin
- Department of Physiology, China Medical University, Taichung 40402, Taiwan
| | - Cing-Yu Chen
- Department of Cosmetic Science, Providence University, Taichung, Taiwan; School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Paul Chan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yuk-Man Leung
- Department of Physiology, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
27
|
Kho C. Targeting calcium regulators as therapy for heart failure: focus on the sarcoplasmic reticulum Ca-ATPase pump. Front Cardiovasc Med 2023; 10:1185261. [PMID: 37534277 PMCID: PMC10392702 DOI: 10.3389/fcvm.2023.1185261] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/06/2023] [Indexed: 08/04/2023] Open
Abstract
Impaired myocardial Ca2+ cycling is a critical contributor to the development of heart failure (HF), causing changes in the contractile function and structure remodeling of the heart. Within cardiomyocytes, the regulation of sarcoplasmic reticulum (SR) Ca2+ storage and release is largely dependent on Ca2+ handling proteins, such as the SR Ca2+ ATPase (SERCA2a) pump. During the relaxation phase of the cardiac cycle (diastole), SERCA2a plays a critical role in transporting cytosolic Ca2+ back to the SR, which helps to restore both cytosolic Ca2+ levels to their resting state and SR Ca2+ content for the next contraction. However, decreased SERCA2a expression and/or pump activity are key features in HF. As a result, there is a growing interest in developing therapeutic approaches to target SERCA2a. This review provides an overview of the regulatory mechanisms of the SERCA2a pump and explores potential strategies for SERCA2a-targeted therapy, which are being investigated in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Changwon Kho
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
28
|
Dahl R, Moore AC, Knight C, Mauger C, Zhang H, Schiltz GE, Koss WA, Bezprozvanny I. Positive Allosteric Modulator of SERCA Pump NDC-1173 Exerts Beneficial Effects in Mouse Model of Alzheimer's Disease. Int J Mol Sci 2023; 24:11057. [PMID: 37446234 PMCID: PMC10341805 DOI: 10.3390/ijms241311057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disease that affects millions of people worldwide. AD does not have a cure and most drug development efforts in the AD field have been focused on targeting the amyloid pathway based on the "amyloid cascade hypothesis". However, in addition to the amyloid pathway, substantial evidence also points to dysregulated neuronal calcium (Ca2+) signaling as one of the key pathogenic events in AD, and it has been proposed that pharmacological agents that stabilize neuronal Ca2+ signaling may act as disease-modifying agents in AD. In previous studies, we demonstrated that positive allosteric regulators (PAMs) of the Sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) pump might act as such Ca2+ stabilizing agents. In the present study, we report the development of a novel SERCA PAM agent, compound NDC-1173. To test the effectiveness of this compound, we performed behavioral studies with the APP/PS1 transgenic AD mouse model. We also evaluated effects of this compound on expression of endoplasmic reticulum (ER) stress genes in the hippocampus of APP/PS1 mice. The results of this study support the hypothesis that the SERCA pump is a potential novel therapeutic drug target and that NDC-1173 is a promising lead molecule for developing disease-modifying agents in AD.
Collapse
Affiliation(s)
- Russell Dahl
- Neurodon, 9800 Connecticut Drive, Crown Point, IN 46307, USA;
| | - Amanda C. Moore
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; (A.C.M.); (W.A.K.)
| | - Caitlynn Knight
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; (C.K.); (H.Z.)
| | - Colleen Mauger
- Neurodon, 9800 Connecticut Drive, Crown Point, IN 46307, USA;
| | - Hua Zhang
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; (C.K.); (H.Z.)
| | - Gary E. Schiltz
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA;
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
| | - Wendy A. Koss
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; (A.C.M.); (W.A.K.)
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA; (C.K.); (H.Z.)
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, 194021 St. Petersburg, Russia
| |
Collapse
|
29
|
Gao Y, Su X, Xue T, Zhang N. The beneficial effects of astragaloside IV on ameliorating diabetic kidney disease. Biomed Pharmacother 2023; 163:114598. [PMID: 37150034 DOI: 10.1016/j.biopha.2023.114598] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/22/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic kidney disease (DKD) has become the major cause of chronic kidney disease or end-stage renal disease. There is still a need for innovative treatment strategies for preventing, arresting, treating, and reversing DKD, and a plethora of scientific evidence has revealed that Chinese herbal monomers can attenuate DKD in multiple ways. Astragaloside IV (AS-IV) is one of the active ingredients of Astragalus membranaceus and was selected as a chemical marker in the Chinese Pharmacopeia for quality control purposes. An increasing amount of studies indicate that AS-IV is a promising novel drug for the treatment of DKD. AS-IV has been shown to improve DKD by combating oxidative stress, attenuating endoplasmic reticulum stress, regulating calcium homeostasis, alleviating inflammation, improving vascular function, improving epithelial to mesenchymal transition and so on. This review briefly summarizes the pathogenesis of DKD, systematically reviews the mechanisms by which AS-IV improves DKD, and aims to facilitate related pharmacological research and development to promote the utilization of Chinese herbal monomers in DKD.
Collapse
Affiliation(s)
- Yiwei Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Xin Su
- Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Taiqi Xue
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China.
| |
Collapse
|
30
|
Yang Y, Xie E, Liu Y, Peng Z, Yu C, Hua K, Yang X. Calcium promotes vascular smooth muscle cell phenotypic switching in Marfan syndrome. Biochem Biophys Res Commun 2023; 665:124-132. [PMID: 37156050 DOI: 10.1016/j.bbrc.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
Fibrillin 1 (Fbn1) mutations cause Marfan syndrome (MFS), with aortic root dilatation, dissection, and rupture. Few studies reported the blood calcium and lipid profile of MFS, and the effect of vascular smooth muscle cell (VSMC) phenotypic switching on MFS aortic aneurysm is unclear. Here, we aimed to investigate the role of calcium-related VSMC phenotypic switching in MFS. We retrospectively collected MFS patients' clinical data, performed bioinformatics analysis to screen the enriched biological process in MFS patients and mice, and detected markers of VSMC phenotypic switching on Fbn1C1039G/+ mice and primary aortic vascular smooth muscle cells. We found that patients with MFS have elevated blood calcium levels and dyslipidemia. Furthermore, the calcium concentration levels were increased with age in MFS mice, accompanied by the promoted VSMC phenotypic switching, and SERCA2 contributed to maintaining the contractile phenotype of VSMCs. This study provides the first evidence that the increased calcium is associated with the promoted VSMC phenotype switching in MFS. SERCA may become a novel therapeutic target for suppressing aneurysm progression in MFS.
Collapse
Affiliation(s)
- Yunxiao Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Enzehua Xie
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100047, China
| | - Yuhua Liu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhan Peng
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Cuntao Yu
- Department of Cardiovascular Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular Diseases, Beijing, 100047, China.
| | - Kun Hua
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Xiubin Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
31
|
Zhang Y, Han S, Liu C, Zheng Y, Li H, Gao F, Bian Y, Liu X, Liu H, Hu S, Li Y, Chen ZJ, Zhao S, Zhao H. THADA inhibition in mice protects against type 2 diabetes mellitus by improving pancreatic β-cell function and preserving β-cell mass. Nat Commun 2023; 14:1020. [PMID: 36823211 PMCID: PMC9950491 DOI: 10.1038/s41467-023-36680-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Impaired insulin secretion is a hallmark in type 2 diabetes mellitus (T2DM). THADA has been identified as a candidate gene for T2DM, but its role in glucose homeostasis remains elusive. Here we report that THADA is strongly activated in human and mouse islets of T2DM. Both global and β-cell-specific Thada-knockout mice exhibit improved glycemic control owing to enhanced β-cell function and decreased β-cell apoptosis. THADA reduces endoplasmic reticulum (ER) Ca2+ stores in β-cells by inhibiting Ca2+ re-uptake via SERCA2 and inducing Ca2+ leakage through RyR2. Upon persistent ER stress, THADA interacts with and activates the pro-apoptotic complex comprising DR5, FADD and caspase-8, thus aggravating ER stress-induced apoptosis. Importantly, THADA deficiency protects mice from high-fat high-sucrose diet- and streptozotocin-induced hyperglycemia by restoring insulin secretion and preserving β-cell mass. Moreover, treatment with alnustone inhibits THADA's function, resulting in ameliorated hyperglycemia in obese mice. Collectively, our results support pursuit of THADA as a potential target for developing T2DM therapies.
Collapse
Affiliation(s)
- Yuqing Zhang
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shan Han
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Congcong Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuanwen Zheng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China
| | - Hao Li
- Shandong Provincial Qianfoshan Hospital, Shandong University, 250014, Jinan, Shandong, China
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Science, 100101, Beijing, China
| | - Yuehong Bian
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Xin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Shourui Hu
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Yuxuan Li
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China. .,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, 200135, Shanghai, China. .,Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, 250012, Jinan, China.
| | - Shigang Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250012, Jinan, Shandong, China.
| |
Collapse
|
32
|
Hunter KD, Crozier RWE, Braun JL, Fajardo VA, MacNeil AJ. Acute activation of SERCA with CDN1163 attenuates IgE-mediated mast cell activation through selective impairment of ROS and p38 signaling. FASEB J 2023; 37:e22748. [PMID: 36624659 DOI: 10.1096/fj.202201272r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/13/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
Mast cells are granulocytic immune sentinels present in vascularized tissues that drive chronic inflammatory mechanisms characteristic of allergic pathologies. IgE-mediated mast cell activation leads to a rapid mobilization of Ca2+ from intracellular stores, which is essential for the release of preformed mediators via degranulation and de novo synthesized proinflammatory cytokines and chemokines. Given its potent signaling capacity, the dynamics of Ca2+ localization are highly regulated by various pumps and channels controlling cytosolic Ca2+ concentrations. Among these is sarco/endoplasmic reticulum Ca2+ -ATPase (SERCA), which functions to maintain low cytosolic Ca2+ concentrations by actively transporting cytosolic Ca2+ ions into the endoplasmic reticulum. In this study, we characterized the role of SERCA in allergen-activated mast cells using IgE-sensitized bone marrow-derived mast cells (BMMCs) treated with the SERCA activating compound, CDN1163, and simultaneously stimulated with allergen through FcεRI under stem cell factor (SCF) potentiation. Acute treatment with CDN1163 was found to attenuate early phase mast cell degranulation along with reactive oxygen species (ROS) production. Additionally, treatment with CDN1163 significantly reduced secretion of IL-6, IL-13, and CCL3, suggesting a role for SERCA in the late phase mast cell response. The protective effects of SERCA activation via CDN1163 treatment on the early and late phase mast cell response may be driven by the selective suppression of p38 MAPK signaling. Together, these findings implicate SERCA as an important regulator of the mast cell response to allergen and suggest SERCA activity may offer therapeutic potential targeting allergic pathologies, warranting further investigation.
Collapse
Affiliation(s)
- Katie D Hunter
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Robert W E Crozier
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Jessica L Braun
- Department of Kinesiology, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Val A Fajardo
- Department of Kinesiology, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Faculty of Applied Health Sciences, Cairns Family Health and Bioscience Research Complex, Brock University, Niagara Region, Ontario, Canada
| |
Collapse
|
33
|
Abstract
Rather than serving as a mere onlooker, adipose tissue is a complex endocrine organ and active participant in disease initiation and progression. Disruptions of biological processes operating within adipose can disturb healthy systemic physiology, the sequelae of which include metabolic disorders such as obesity and type 2 diabetes. A burgeoning interest in the field of adipose research has allowed for the elucidation of regulatory networks underlying both adipose tissue function and dysfunction. Despite this progress, few diseases are treated by targeting maladaptation in the adipose, an oft-overlooked organ. In this review, we elaborate on the distinct subtypes of adipocytes, their developmental origins and secretory roles, and the dynamic interplay at work within the tissue itself. Central to this discussion is the relationship between adipose and disease states, including obesity, cachexia, and infectious diseases, as we aim to leverage our wealth of knowledge for the development of novel and targeted therapeutics.
Collapse
Affiliation(s)
- Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; .,Howard Hughes Medical Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
34
|
Nikolaienko R, Bovo E, Yuen SL, Treinen LM, Berg K, Aldrich CC, Thomas DD, Cornea RL, Zima AV. New N-aryl-N-alkyl-thiophene-2-carboxamide compound enhances intracellular Ca 2+ dynamics by increasing SERCA2a Ca 2+ pumping. Biophys J 2023; 122:386-396. [PMID: 36463408 PMCID: PMC9892616 DOI: 10.1016/j.bpj.2022.12.002] [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: 07/12/2022] [Revised: 10/31/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The type 2a sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) plays a central role in the intracellular Ca2+ homeostasis of cardiac myocytes, pumping Ca2+ from the cytoplasm into the sarcoplasmic reticulum (SR) lumen to maintain relaxation (diastole) and prepare for contraction (systole). Diminished SERCA2a function has been reported in several pathological conditions, including heart failure. Therefore, development of new drugs that improve SERCA2a Ca2+ transport is of great clinical significance. In this study, we characterized the effect of a recently identified N-aryl-N-alkyl-thiophene-2-carboxamide (or compound 1) on SERCA2a Ca2+-ATPase and Ca2+ transport activities in cardiac SR vesicles, and on Ca2+ regulation in a HEK293 cell expression system and in mouse ventricular myocytes. We found that compound 1 enhances SERCA2a Ca2+-ATPase and Ca2+ transport in SR vesicles. Fluorescence lifetime measurements of fluorescence resonance energy transfer between SERCA2a and phospholamban indicated that compound 1 interacts with the SERCA-phospholamban complex. Measurement of endoplasmic reticulum Ca2+ dynamics in HEK293 cells expressing human SERCA2a showed that compound 1 increases endoplasmic reticulum Ca2+ load by enhancing SERCA2a-mediated Ca2+ transport. Analysis of cytosolic Ca2+ dynamics in mouse ventricular myocytes revealed that compound 1 increases the action potential-induced Ca2+ transients and SR Ca2+ load, with negligible effects on L-type Ca2+ channels and Na+/Ca2+ exchanger. However, during adrenergic receptor activation, compound 1 did not further increase Ca2+ transients and SR Ca2+ load, but it decreased the propensity toward Ca2+ waves. Suggestive of concurrent desirable effects of compound 1 on RyR2, [3H]-ryanodine binding to cardiac SR vesicles shows a small decrease in nM Ca2+ and a small increase in μM Ca2+. Accordingly, compound 1 slightly decreased Ca2+ sparks in permeabilized myocytes. Thus, this novel compound shows promising characteristics to improve intracellular Ca2+ dynamics in cardiomyocytes that exhibit reduced SERCA2a Ca2+ uptake, as found in failing hearts.
Collapse
Affiliation(s)
- Roman Nikolaienko
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Samantha L Yuen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Levy M Treinen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Kaja Berg
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Courtney C Aldrich
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois.
| |
Collapse
|
35
|
Curcumin Stimulates UCP1-independent Thermogenesis in 3T3-L1 White Adipocytes but Suppresses in C2C12 Muscle Cells. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-022-0319-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
36
|
A Large-Scale High-Throughput Screen for Modulators of SERCA Activity. Biomolecules 2022; 12:biom12121789. [PMID: 36551215 PMCID: PMC9776381 DOI: 10.3390/biom12121789] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The sarco/endoplasmic reticulum Ca-ATPase (SERCA) is a P-type ion pump that transports Ca2+ from the cytosol into the endoplasmic/sarcoplasmic reticulum (ER/SR) in most mammalian cells. It is critically important in muscle, facilitating relaxation and enabling subsequent contraction. Increasing SERCA expression or specific activity can alleviate muscle dysfunction, most notably in the heart, and we seek to develop small-molecule drug candidates that activate SERCA. Therefore, we adapted an NADH-coupled assay, measuring Ca-dependent ATPase activity of SERCA, to high-throughput screening (HTS) format, and screened a 46,000-compound library of diverse chemical scaffolds. This HTS platform yielded numerous hits that reproducibly alter SERCA Ca-ATPase activity, with few false positives. The top 19 activating hits were further tested for effects on both Ca-ATPase and Ca2+ transport, in both cardiac and skeletal SR. Nearly all hits increased Ca2+ uptake in both cardiac and skeletal SR, with some showing isoform specificity. Furthermore, dual analysis of both activities identified compounds with a range of effects on Ca2+-uptake and ATPase, which fit into distinct classifications. Further study will be needed to identify which classifications are best suited for therapeutic use. These results reinforce the need for robust secondary assays and criteria for selection of lead compounds, before undergoing HTS on a larger scale.
Collapse
|
37
|
Hricovíni M, Owens RJ, Bak A, Kozik V, Musiał W, Pierattelli R, Májeková M, Rodríguez Y, Musioł R, Slodek A, Štarha P, Piętak K, Słota D, Florkiewicz W, Sobczak-Kupiec A, Jampílek J. Chemistry towards Biology-Instruct: Snapshot. Int J Mol Sci 2022; 23:14815. [PMID: 36499140 PMCID: PMC9739621 DOI: 10.3390/ijms232314815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
The knowledge of interactions between different molecules is undoubtedly the driving force of all contemporary biomedical and biological sciences. Chemical biology/biological chemistry has become an important multidisciplinary bridge connecting the perspectives of chemistry and biology to the study of small molecules/peptidomimetics and their interactions in biological systems. Advances in structural biology research, in particular linking atomic structure to molecular properties and cellular context, are essential for the sophisticated design of new medicines that exhibit a high degree of druggability and very importantly, druglikeness. The authors of this contribution are outstanding scientists in the field who provided a brief overview of their work, which is arranged from in silico investigation through the characterization of interactions of compounds with biomolecules to bioactive materials.
Collapse
Affiliation(s)
- Miloš Hricovíni
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 845 38 Bratislava, Slovakia
| | - Raymond J. Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK, University of Oxford, Oxford OX11 0QS, UK
- Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Violetta Kozik
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Pharmaceutical Faculty, Wroclaw Medical University, Borowska 211A, 50 556 Wrocław, Poland
| | - Roberta Pierattelli
- Magnetic Resonance Center and Department of Chemistry “Ugo Schiff”, University of Florence, 50019 Sesto Fiorentino, Italy
| | - Magdaléna Májeková
- Center of Experimental Medicine SAS and Department of Biochemical Pharmacology, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovakia
| | - Yoel Rodríguez
- Department of Natural Sciences, Eugenio María de Hostos Community College, City University of New York, 500 Grand Concourse, Bronx, NY 10451, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Robert Musioł
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Aneta Slodek
- Institute of Chemistry, University of Silesia, Szkolna 9, 40 007 Katowice, Poland
| | - Pavel Štarha
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 771 46 Olomouc, Czech Republic
| | - Karina Piętak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Dagmara Słota
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Wioletta Florkiewicz
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Agnieszka Sobczak-Kupiec
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31 864 Krakow, Poland
| | - Josef Jampílek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
38
|
Badin J, Rodenbeck S, McKenney-Drake ML, Sturek M. Multiphasic changes in smooth muscle Ca 2+ transporters during the progression of coronary atherosclerosis. CURRENT TOPICS IN MEMBRANES 2022; 90:95-121. [PMID: 36368876 DOI: 10.1016/bs.ctm.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Ischemic heart disease due to macrovascular atherosclerosis and microvascular dysfunction is the major cause of death worldwide and the unabated increase in metabolic syndrome is a major reason why this will continue. Intracellular free Ca2+ ([Ca2+]i) regulates a variety of cellular functions including contraction, proliferation, migration, and transcription. It follows that studies of vascular Ca2+ regulation in reductionist models and translational animal models are vital to understanding vascular health and disease. Swine with metabolic syndrome (MetS) develop the full range of coronary atherosclerosis from mild to severe disease. Intravascular imaging enables quantitative measurement of atherosclerosis in vivo, so viable coronary smooth muscle (CSM) cells can be dispersed from the arteries to enable Ca2+ transport studies in native cells. Transition of CSM from the contractile phenotype in the healthy swine to the proliferative phenotype in mild atherosclerosis was associated with increases in SERCA activity, sarcoplasmic reticulum Ca2+, and voltage-gated Ca2+ channel function. In vitro organ culture confirmed that SERCA activation induces CSM proliferation. Transition from the proliferative to a more osteogenic phenotype was associated with decreases in all three Ca2+ transporters. Overall, there was a biphasic change in Ca2+ transporters over the progression of atherosclerosis in the swine model and this was confirmed in CSM from failing explanted hearts of humans. A major determinant of endolysosome content in human CSM is the severity of atherosclerosis. In swine CSM endolysosome Ca2+ release occurred through the TPC2 channel. We propose a multiphasic change in Ca2+ transporters over the progression of coronary atherosclerosis.
Collapse
Affiliation(s)
- Jill Badin
- ZOLL Medical Corporation, Chelmsford, MA, United States
| | - Stacey Rodenbeck
- Department of Biology, Harding University, Searcy, AR, United States
| | - Mikaela L McKenney-Drake
- Butler University, Health Sciences Department, Pharmacy and Health Sciences, Indianapolis, IN, United States
| | - Michael Sturek
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
39
|
Suppression of Ca 2+ oscillations by SERCA inhibition in human alveolar type 2 A549 cells: rescue by ochratoxin A but not CDN1163. Life Sci 2022; 308:120913. [PMID: 36037871 DOI: 10.1016/j.lfs.2022.120913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022]
Abstract
AIMS Lung type 2 alveolar cells, by secreting surfactant to lower surface tension, contribute to enhance lung compliance. Stretching, as a result of lung expansion, triggers type 1 alveolar cell to release ATP, which in turn stimulates Ca2+-dependent surfactant secretion by neighboring type 2 cells. In this report, we studied ATP-triggered Ca2+ signaling in human alveolar type 2 A549 cells. MAIN METHODS Ca2+ signaling was examined using microfluorimetric measurement with fura-2 as fluorescent dye. KEY FINDINGS Ca2+ oscillations triggered by ATP relied on inositol 1,4,5-trisphosphate-induced Ca2+ release and store-operated Ca2+ entry. Pathological conditions such as influenza virus infection and diabetes reportedly inhibit sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA). We found that a very mild inhibition of SERCA by cyclopiazonic acid (CPA) sufficed to decrease Ca2+ oscillation frequency and the percentage of cells exhibiting Ca2+ oscillations. Ochratoxin A (OTA), an activator of SERCA, could prevent the suppressive effects by CPA. Inhibition of SERCA by hydrogen peroxide also suppressed Ca2+ oscillations. Interestingly, hydrogen peroxide-induced inhibition was prevented by OTA but aggravated by CDN1163, an allosteric activator of SERCA. CDN1163 also had an untoward effect of releasing intracellular Ca2+. SIGNIFICANCE Different modes of activation of SERCA may determine the outcome of rescue of Ca2+ oscillations in case of SERCA inhibition in alveolar type 2 cells.
Collapse
|
40
|
Natural Polyphenols as SERCA Activators: Role in the Endoplasmic Reticulum Stress-Related Diseases. Molecules 2022; 27:molecules27165095. [PMID: 36014327 PMCID: PMC9415898 DOI: 10.3390/molecules27165095] [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: 06/16/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) is a key protein responsible for transporting Ca2+ ions from the cytosol into the lumen of the sarco/endoplasmic reticulum (SR/ER), thus maintaining Ca2+ homeostasis within cells. Accumulating evidence suggests that impaired SERCA function is associated with disruption of intracellular Ca2+ homeostasis and induction of ER stress, leading to different chronic pathological conditions. Therefore, appropriate strategies to control Ca2+ homeostasis via modulation of either SERCA pump activity/expression or relevant signaling pathways may represent a useful approach to combat pathological states associated with ER stress. Natural dietary polyphenolic compounds, such as resveratrol, gingerol, ellagic acid, luteolin, or green tea polyphenols, with a number of health-promoting properties, have been described either to increase SERCA activity/expression directly or to affect Ca2+ signaling pathways. In this review, potential Ca2+-mediated effects of the most studied polyphenols on SERCA pumps or related Ca2+ signaling pathways are summarized, and relevant mechanisms of their action on Ca2+ regulation with respect to various ER stress-related states are depicted. All data were collected using scientific search tools (i.e., Science Direct, PubMed, Scopus, and Google Scholar).
Collapse
|
41
|
Huang X, Lin X, Wang L, Xie Y, Que Y, Li S, Hu P, Tong X. Substitution of SERCA2 Cys 674 aggravates cardiac fibrosis by promoting the transformation of cardiac fibroblasts to cardiac myofibroblasts. Biochem Pharmacol 2022; 203:115164. [PMID: 35809651 DOI: 10.1016/j.bcp.2022.115164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/02/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) is vital to maintain intracellular calcium homeostasis, and its redox Cys674 (C674) is the key to regulating activity. Our goal was to investigate whether the redox state of SERCA2 C674 is critical for cardiac fibrosis and the mechanisms involved. Heterozygous SERCA2 C674S knock-in (SKI) mice, in which half of C674 was substituted by serine, were used to mimic the partial loss of the reactive C674 thiol in pathological conditions. In cardiac fibroblasts, the substitution of C674 thiol increased Ca2+ levels in cytoplasm and mitochondria, and intracellular ROS levels, and activated calcineurin/nuclear factor of activated T-lymphocytes (NFAT) pathway, increased the protein expression of profibrotic factors TGF beta 1 (TGF-β1), alpha smooth muscle actin, collagen I and collagen III, and promoted the transformation of cardiac fibroblasts to cardiac myofibroblasts, which could be reversed by calcineurin/NFAT inhibitor, SERCA2 agonist, or ROS scavenger. Activation of SERCA2 or scavenging ROS is beneficial to alleviate cardiac fibrosis caused by the substitution of C674. In conclusion, the partial loss of the reactive C674 thiol in the SERCA2 exacerbates cardiac fibrosis by activating the calcineurin/NFAT/TGF-β1 pathway to promote the transformation of cardiac fibroblasts to cardiac myofibroblasts, which highlights the importance of C674 redox state in maintaining the homeostasis of cardiac fibroblasts. SERCA2 is a potential therapeutic target for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Xiaoyang Huang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaojuan Lin
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Langtao Wang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yufei Xie
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
42
|
Bauzá-Thorbrügge M, Banke E, Chanclón B, Peris E, Wu Y, Musovic S, Jönsson C, Strålfors P, Rorsman P, Olofsson CS, Asterholm IW. Adipocyte-specific ablation of the Ca 2+ pump SERCA2 impairs whole-body metabolic function and reveals the diverse metabolic flexibility of white and brown adipose tissue. Mol Metab 2022; 63:101535. [PMID: 35760318 PMCID: PMC9287368 DOI: 10.1016/j.molmet.2022.101535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) transports Ca2+ from the cytosol into the ER and is essential for appropriate regulation of intracellular Ca2+ homeostasis. The objective of this study was to test the hypothesis that SERCA pumps are involved in the regulation of white adipocyte hormone secretion and other aspects of adipose tissue function and that this control is disturbed in obesity-induced type-2 diabetes. METHODS SERCA expression was measured in isolated human and mouse adipocytes as well as in whole mouse adipose tissue by Western blot and RT-qPCR. To test the significance of SERCA2 in adipocyte functionality and whole-body metabolism, we generated adipocyte-specific SERCA2 knockout mice. The mice were metabolically phenotyped by glucose tolerance and tracer studies, histological analyses, measurements of glucose-stimulated insulin release in isolated islets, and gene/protein expression analyses. We also tested the effect of pharmacological SERCA inhibition and genetic SERCA2 ablation in cultured adipocytes. Intracellular and mitochondrial Ca2+ levels were recorded with dual-wavelength ratio imaging and mitochondrial function was assessed by Seahorse technology. RESULTS We demonstrate that SERCA2 is downregulated in white adipocytes from patients with obesity and type-2 diabetes as well as in adipocytes from diet-induced obese mice. SERCA2-ablated adipocytes display disturbed Ca2+ homeostasis associated with upregulated ER stress markers and impaired hormone release. These adipocyte alterations are linked to mild lipodystrophy, reduced adiponectin levels, and impaired glucose tolerance. Interestingly, adipocyte-specific SERCA2 ablation leads to increased glucose uptake in white adipose tissue while glucose uptake is reduced in brown adipose tissue. This dichotomous effect on glucose uptake is due to differently regulated mitochondrial function. In white adipocytes, SERCA2 deficiency triggers an adaptive increase in FGF21, increased mitochondrial UCP1 levels, and increased oxygen consumption rate (OCR). In contrast, brown SERCA2 null adipocytes display reduced OCR despite increased mitochondrial content and UCP1 levels compared to wild type controls. CONCLUSIONS Our data suggest causal links between reduced white adipocyte SERCA2 levels, deranged adipocyte Ca2+ homeostasis, adipose tissue dysfunction and type-2 diabetes.
Collapse
Affiliation(s)
- Marco Bauzá-Thorbrügge
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Elin Banke
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Belén Chanclón
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Eduard Peris
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Yanling Wu
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Saliha Musovic
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Cecilia Jönsson
- Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden.
| | - Peter Strålfors
- Department of Biomedical and Clinical Sciences, Linköping University, SE-58185 Linköping, Sweden.
| | - Patrik Rorsman
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden; Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX4 7LE, UK.
| | - Charlotta S Olofsson
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 11, SE-405 30 Göteborg, Sweden.
| |
Collapse
|
43
|
Kimura T, Kagami K, Sato A, Osaki A, Ito K, Horii S, Toya T, Masaki N, Yasuda R, Nagatomo Y, Adachi T. Sarco/Endoplasmic Reticulum Ca 2+ ATPase 2 Activator Ameliorates Endothelial Dysfunction; Insulin Resistance in Diabetic Mice. Cells 2022; 11:1488. [PMID: 35563793 PMCID: PMC9099866 DOI: 10.3390/cells11091488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Sarco/endoplasmic reticulum Ca2+-ATPase2 (SERCA2) is impaired in various organs in animal models of diabetes. The purpose of this study was to test the effects of an allosteric SERCA2 activator (CDN1163) on glucose intolerance, hepatosteatosis, skeletal muscle function, and endothelial dysfunction in diabetic (db/db) mice. Methods: Either CDN1163 or vehicle was injected intraperitoneally into 16-week-old male control and db/db mice for 5 consecutive days. Results: SERCA2 protein expression was decreased in the aorta of db/db mice. In isometric tension measurements of aortic rings from db/db mice treated with CDN1163, acetylcholine (ACh)-induced relaxation was improved. In vivo intraperitoneal administrations of CDN 1163 also increased ACh-induced relaxation. Moreover, CDN1163 significantly decreased blood glucose in db/db mice at 60 and 120 min during a glucose tolerance test; it also decreased serum insulin levels, hepatosteatosis, and oxygen consumption in skeletal muscle during the early period of exercise in db/db mice. Conclusions: CDN1163 directly improved aortic endothelial dysfunction in db/db mice. Moreover, CDN1163 improved hepatosteatosis, skeletal muscle function, and insulin resistance in db/db mice. The activation of SERCA2 might be a strategy for the all the tissue expressed SERCA2a improvement of endothelial dysfunction and the target for the organs related to insulin resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yuji Nagatomo
- Department of Internal Medicine I, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan; (T.K.); (K.K.); (A.S.); (A.O.); (K.I.); (S.H.); (T.T.); (N.M.); (R.Y.)
| | - Takeshi Adachi
- Department of Internal Medicine I, Division of Cardiovascular Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Japan; (T.K.); (K.K.); (A.S.); (A.O.); (K.I.); (S.H.); (T.T.); (N.M.); (R.Y.)
| |
Collapse
|
44
|
Lee Y, Zawieja SD, Muthuchamy M. Lymphatic Collecting Vessel: New Perspectives on Mechanisms of Contractile Regulation and Potential Lymphatic Contractile Pathways to Target in Obesity and Metabolic Diseases. Front Pharmacol 2022; 13:848088. [PMID: 35355722 PMCID: PMC8959455 DOI: 10.3389/fphar.2022.848088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 01/19/2023] Open
Abstract
Obesity and metabolic syndrome pose a significant risk for developing cardiovascular disease and remain a critical healthcare challenge. Given the lymphatic system's role as a nexus for lipid absorption, immune cell trafficking, interstitial fluid and macromolecule homeostasis maintenance, the impact of obesity and metabolic disease on lymphatic function is a burgeoning field in lymphatic research. Work over the past decade has progressed from the association of an obese phenotype with Prox1 haploinsufficiency and the identification of obesity as a risk factor for lymphedema to consistent findings of lymphatic collecting vessel dysfunction across multiple metabolic disease models and organisms and characterization of obesity-induced lymphedema in the morbidly obese. Critically, recent findings have suggested that restoration of lymphatic function can also ameliorate obesity and insulin resistance, positing lymphatic targeted therapies as relevant pharmacological interventions. There remain, however, significant gaps in our understanding of lymphatic collecting vessel function, particularly the mechanisms that regulate the spontaneous contractile activity required for active lymph propulsion and lymph return in humans. In this article, we will review the current findings on lymphatic architecture and collecting vessel function, including recent advances in the ionic basis of lymphatic muscle contractile activity. We will then discuss lymphatic dysfunction observed with metabolic disruption and potential pathways to target with pharmacological approaches to improve lymphatic collecting vessel function.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Scott D Zawieja
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
45
|
Lasa-Elgarresta J, Mosqueira-Martín L, González-Imaz K, Marco-Moreno P, Gerenu G, Mamchaoui K, Mouly V, López de Munain A, Vallejo-Illarramendi A. Targeting the Ubiquitin-Proteasome System in Limb-Girdle Muscular Dystrophy With CAPN3 Mutations. Front Cell Dev Biol 2022; 10:822563. [PMID: 35309930 PMCID: PMC8924035 DOI: 10.3389/fcell.2022.822563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
LGMDR1 is caused by mutations in the CAPN3 gene that encodes calpain 3 (CAPN3), a non-lysosomal cysteine protease necessary for proper muscle function. Our previous findings show that CAPN3 deficiency leads to reduced SERCA levels through increased protein degradation. This work investigates the potential contribution of the ubiquitin-proteasome pathway to increased SERCA degradation in LGMDR1. Consistent with our previous results, we observed that CAPN3-deficient human myotubes exhibit reduced SERCA protein levels and high cytosolic calcium concentration. Treatment with the proteasome inhibitor bortezomib (Velcade) increased SERCA2 protein levels and normalized intracellular calcium levels in CAPN3-deficient myotubes. Moreover, bortezomib was able to recover mutated CAPN3 protein in a patient carrying R289W and R546L missense mutations. We found that CAPN3 knockout mice (C3KO) presented SERCA deficits in skeletal muscle in the early stages of the disease, prior to the manifestation of muscle deficits. However, treatment with bortezomib (0.8 mg/kg every 72 h) for 3 weeks did not rescue SERCA levels. No change in muscle proteasome activity was observed in bortezomib-treated animals, suggesting that higher bortezomib doses are needed to rescue SERCA levels in this model. Overall, our results lay the foundation for exploring inhibition of the ubiquitin-proteasome as a new therapeutic target to treat LGMDR1 patients. Moreover, patients carrying missense mutations in CAPN3 and presumably other genes may benefit from proteasome inhibition by rescuing mutant protein levels. Further studies in suitable models will be necessary to demonstrate the therapeutic efficacy of proteasome inhibition for different missense mutations.
Collapse
Affiliation(s)
- Jaione Lasa-Elgarresta
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Laura Mosqueira-Martín
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Klaudia González-Imaz
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Pablo Marco-Moreno
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Gorka Gerenu
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Department of Physiology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Adolfo López de Munain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| |
Collapse
|
46
|
Lebeau PF, Byun JH, Platko K, Saliba P, Sguazzin M, MacDonald ME, Paré G, Steinberg GR, Janssen LJ, Igdoura SA, Tarnopolsky MA, Wayne Chen SR, Seidah NG, Magolan J, Austin RC. Caffeine blocks SREBP2-induced hepatic PCSK9 expression to enhance LDLR-mediated cholesterol clearance. Nat Commun 2022; 13:770. [PMID: 35140212 PMCID: PMC8828868 DOI: 10.1038/s41467-022-28240-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/05/2022] [Indexed: 01/06/2023] Open
Abstract
Evidence suggests that caffeine (CF) reduces cardiovascular disease (CVD) risk. However, the mechanism by which this occurs has not yet been uncovered. Here, we investigated the effect of CF on the expression of two bona fide regulators of circulating low-density lipoprotein cholesterol (LDLc) levels; the proprotein convertase subtilisin/kexin type 9 (PCSK9) and the low-density lipoprotein receptor (LDLR). Following the observation that CF reduced circulating PCSK9 levels and increased hepatic LDLR expression, additional CF-derived analogs with increased potency for PCSK9 inhibition compared to CF itself were developed. The PCSK9-lowering effect of CF was subsequently confirmed in a cohort of healthy volunteers. Mechanistically, we demonstrate that CF increases hepatic endoplasmic reticulum (ER) Ca2+ levels to block transcriptional activation of the sterol regulatory element-binding protein 2 (SREBP2) responsible for the regulation of PCSK9, thereby increasing the expression of the LDLR and clearance of LDLc. Our findings highlight ER Ca2+ as a master regulator of cholesterol metabolism and identify a mechanism by which CF may protect against CVD.
Collapse
Affiliation(s)
- Paul F Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, L8N 4A6, Canada
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, L8N 4A6, Canada
| | - Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, L8N 4A6, Canada
| | - Paul Saliba
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Matthew Sguazzin
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Melissa E MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, L8N 4A6, Canada
| | - Guillaume Paré
- Population Health Research Institute, McMaster University, Hamilton, ON, L8L 2X2, Canada.,The Departments of Medicine, Epidemiology and Pathology, McMaster University, Hamilton, ON, L8L 2X2, Canada.,The Thrombosis and Atherosclerosis Research Institute (TaARI), Department of Medicine, David Braley Research Institute, McMaster University, Hamilton, L8L 2X2, Canada
| | - Gregory R Steinberg
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4L8, Canada.,Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Luke J Janssen
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, Hamilton, ON, L8S 4K1, Canada
| | - Suleiman A Igdoura
- Department of Biology and Pathology, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Mark A Tarnopolsky
- Department of Medicine/Neurology, McMaster University, Hamilton, ON, L8N 3Z5, Canada.,Department of Pediatrics, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute of Alberta, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 2T9, Canada
| | - Nabil G Seidah
- Laboratory of Biochemical Neuroendocrinology, Clinical Research Institute of Montreal, affiliated to the University of Montreal, Montreal, QC, H2W 1R7, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, The Research Institute of St. Joe's Hamilton and the Hamilton Center for Kidney Research, Hamilton, ON, L8N 4A6, Canada. .,The Thrombosis and Atherosclerosis Research Institute (TaARI), Department of Medicine, David Braley Research Institute, McMaster University, Hamilton, L8L 2X2, Canada.
| |
Collapse
|
47
|
Zhang D, Niu S, Ma Y, Chen H, Wen Y, Li M, Zhou B, Deng Y, Shi C, Pu G, Yang M, Wang X, Zou C, Chen Y, Ma L. Fenofibrate Improves Insulin Resistance and Hepatic Steatosis and Regulates the Let-7/SERCA2b Axis in High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease Mice. Front Pharmacol 2022; 12:770652. [PMID: 35126113 PMCID: PMC8807641 DOI: 10.3389/fphar.2021.770652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022] Open
Abstract
Fenofibrate is widely used in clinical therapy to effectively ameliorate the development of non-alcoholic fatty liver disease (NAFLD); however, its specific molecular mechanism of action remains largely unknown. MicroRNAs (miRNAs) are key mediators in regulating endoplasmic reticulum (ER) stress during NAFLD, and the deregulation of miRNAs has been demonstrated in NAFLD pathophysiology. The present study aimed to identify whether fenofibrate could influence miRNA expression in NAFLD and investigate the specific mechanism of action of fenofibrate in lipid metabolism disorder-associated diseases. We found that fenofibrate alleviated ER stress and increased the levels of SERCA2b, which serves as a regulator of ER stress. Additionally, the levels of let-7 miRNA were regulated by fenofibrate; let-7 was found to target the 3′ untranslated region of SERCA2b. The present data suggest that the protective effects of fenofibrate against insulin resistance and its suppressive activity against excessive hepatic lipid accumulation may be related to the alteration of the let-7/SERCA2b axis and alleviation of ER stress.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Shanzhuang Niu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yicheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Hang Chen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yu Wen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Mingke Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Bo Zhou
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yi Deng
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chunjing Shi
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Guangyu Pu
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Meng Yang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Xianmei Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chenggang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuanli Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, China
- *Correspondence: Yuanli Chen, ; Lanqing Ma,
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
- *Correspondence: Yuanli Chen, ; Lanqing Ma,
| |
Collapse
|
48
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Chen CL, Huang KT. Emerging Roles of Calcium Signaling in the Development of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2021; 23:ijms23010256. [PMID: 35008682 PMCID: PMC8745268 DOI: 10.3390/ijms23010256] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 01/10/2023] Open
Abstract
The liver plays a central role in energy metabolism. Dysregulated hepatic lipid metabolism is a major cause of non-alcoholic fatty liver disease (NAFLD), a chronic liver disorder closely linked to obesity and insulin resistance. NAFLD is rapidly emerging as a global health problem with currently no approved therapy. While early stages of NAFLD are often considered benign, the disease can progress to an advanced stage that involves chronic inflammation, with increased risk for developing end-stage disease including fibrosis and liver cancer. Hence, there is an urgent need to identify potential pharmacological targets. Ca2+ is an essential signaling molecule involved in a myriad of cellular processes. Intracellular Ca2+ is intricately compartmentalized, and the Ca2+ flow is tightly controlled by a network of Ca2+ transport and buffering proteins. Impaired Ca2+ signaling is strongly associated with endoplasmic reticulum stress, mitochondrial dysfunction and autophagic defects, all of which are etiological factors of NAFLD. In this review, we describe the recent advances that underscore the critical role of dysregulated Ca2+ homeostasis in lipid metabolic abnormalities and discuss the feasibility of targeting Ca2+ signaling as a potential therapeutic approach.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Li-Wen Hsu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Den Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chao-Long Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Tzu Huang
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8193)
| |
Collapse
|
49
|
Mengeste AM, Lund J, Katare P, Ghobadi R, Bakke HG, Lunde PK, Eide L, Mahony GO, Göpel S, Peng XR, Kase ET, Thoresen GH, Rustan AC. The small molecule SERCA activator CDN1163 increases energy metabolism in human skeletal muscle cells. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100060. [PMID: 34909682 PMCID: PMC8663964 DOI: 10.1016/j.crphar.2021.100060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Background and objective A number of studies have highlighted muscle-specific mechanisms of thermogenesis involving futile cycling of Ca2+ driven by sarco (endo)plasmic reticulum Ca2+-ATPase (SERCA) and generating heat from ATP hydrolysis to be a promising strategy to counteract obesity and metabolic dysfunction. However, to the best of our knowledge, no experimental studies concerning the metabolic effects of pharmacologically targeting SERCA in human skeletal muscle cells have been reported. Thus, in the present study, we aimed to explore the effects of SERCA-activating compound, CDN1163, on energy metabolism in differentiated human skeletal muscle cells (myotubes). Methods In this study, we used primary myotube cultures derived from muscle biopsies of the musculus vastus lateralis and musculi interspinales from lean, healthy male donors. Energy metabolism in myotubes was studied using radioactive substrates. Oxygen consumption rate was assessed with the Seahorse XF24 bioanalyzer, whereas metabolic genes and protein expressions were determined by qPCR and immunoblotting, respectively. Results Both acute (4 h) and chronic (5 days) treatment of myotubes with CDN1163 showed increased uptake and oxidation of glucose, as well as complete fatty acid oxidation in the presence of carbonyl cyanide 4-(trifluromethoxy)phenylhydrazone (FCCP). These effects were supported by measurement of oxygen consumption rate, in which the oxidative spare capacity and maximal respiration were enhanced after CDN1163-treatment. In addition, chronic treatment with CDN1163 improved cellular uptake of oleic acid (OA) and fatty acid β-oxidation. The increased OA metabolism was accompanied by enhanced mRNA-expression of carnitine palmitoyl transferase (CPT) 1B, pyruvate dehydrogenase kinase (PDK) 4, as well as increased AMP-activated protein kinase (AMPK)Thr172 phosphorylation. Moreover, following chronic CDN1163 treatment, the expression levels of stearoyl-CoA desaturase (SCD) 1 was decreased together with de novo lipogenesis from acetic acid and formation of diacylglycerol (DAG) from OA. Conclusion Altogether, these results suggest that SERCA activation by CDN1163 enhances energy metabolism in human myotubes, which might be favourable in relation to disorders that are related to metabolic dysfunction such as obesity and type 2 diabetes mellitus. CDN1163 induced an increase in glucose and fatty acid metabolism in primary human myotubes. Myotubes treated with CDN1163 showed lower intramyocellular lipid accumulation and higher rate of β-oxidation. AMPK activity was upregulated in CDN1163-treated myotubes.
Collapse
Key Words
- AMPK
- AMPK, AMP-activated protein kinase
- ASM, acid-soluble metabolites
- CE, cholesteryl ester
- DAG, diacylglycerol
- FA, fatty acid
- FCCP, 4-(trifluromethoxy)phenylhydrazone
- Glucose metabolism
- Lipid metabolism
- OA, oleic acid
- OCR, oxygen consumption rate
- Obesity
- SCD1, stearoyl-CoA desaturase 1
- SERCA
- SERCA, sarco(endo)plasmic reticulum Ca2+-ATPase
- Skeletal muscle
- T2DM, type 2 diabetes mellitus
- Type 2 diabetes
Collapse
Affiliation(s)
- Abel M Mengeste
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Parmeshwar Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Roya Ghobadi
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Hege G Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Norway
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, Norway
| | - Gavin O' Mahony
- Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sven Göpel
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Norway
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| |
Collapse
|
50
|
Guney E, Arruda AP, Parlakgul G, Cagampan E, Min N, Lee GY, Greene L, Tsaousidou E, Inouye K, Han MS, Davis RJ, Hotamisligil GS. Aberrant Ca 2+ signaling by IP 3Rs in adipocytes links inflammation to metabolic dysregulation in obesity. Sci Signal 2021; 14:eabf2059. [PMID: 34905386 PMCID: PMC10130146 DOI: 10.1126/scisignal.abf2059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic metabolic inflammation is a key feature of obesity, insulin resistance, and diabetes. Here, we showed that altered regulation of the Ca2+ channel inositol trisphosphate receptor (IP3R) was an adipocyte-intrinsic event involved in the emergence and propagation of inflammatory signaling and the resulting insulin resistance. Inflammation induced by cytokine exposure in vitro or by obesity in vivo led to increases in the abundance and activity of IP3Rs and in the phosphorylation of the Ca2+-dependent kinase CaMKII in adipocytes in a manner dependent on the kinase JNK. In mice, adipocyte-specific loss of IP3R1/2 protected against adipose tissue inflammation and insulin resistance, despite the mice exhibiting substantial diet-induced weight gain. Thus, this work suggests that increased IP3R activity is a key link between obesity, inflammation, and insulin resistance. These data also suggest that approaches to target IP3R-mediated Ca2+ homeostasis in adipocytes may offer new therapeutic opportunities against metabolic diseases, especially because GWAS studies also implicate this locus in human obesity.
Collapse
Affiliation(s)
- Ekin Guney
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ana Paula Arruda
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Günes Parlakgul
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Erika Cagampan
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Nina Min
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Grace Yankun Lee
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lily Greene
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Eva Tsaousidou
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Karen Inouye
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Myoung Sook Han
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|