1
|
Ma Y, Wang Y, Zhao X, Jin G, Xu J, Li Z, Yin N, Gao Z, Xia B, Peng M. TMEM41B is an endoplasmic reticulum Ca 2+ release channel maintaining naive T cell quiescence and responsiveness. Cell Discov 2025; 11:18. [PMID: 40038246 DOI: 10.1038/s41421-024-00766-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/26/2024] [Indexed: 03/06/2025] Open
Abstract
In mammalian cells, endoplasmic reticulum (ER) passively releases Ca2+ under steady state, but channels involved remain elusive. Here, we report that TMEM41B, an ER-resident membrane protein critical for autophagy, lipid metabolism, and viral infection, functions as an ER Ca2+ release channel. Biochemically, purified recombinant TMEM41B forms a concentration-dependent Ca2+ channel in single-channel electrophysiology assays. Cellularly, TMEM41B deficiency causes ER Ca2+ overload, while overexpression of TMEM41B depletes ER Ca2+. Immunologically, ER Ca2+ overload leads to upregulation of IL-2 and IL-7 receptors in naive T cells, which in turn increases basal signaling of JAK-STAT, AKT-mTOR, and MAPK pathways. This dysregulation drives TMEM41B-deficient naive T cells into a metabolically activated yet immunologically naive state. ER Ca2+ overload also downregulates CD5, lowering the activation threshold of TMEM41B-deficient T cells and leading to heightened T cell responses during infections. In summary, we identify TMEM41B as a concentration-dependent ER Ca2+ release channel, revealing an unexpected role of ER Ca2+ in naive T cell quiescence and responsiveness.
Collapse
Affiliation(s)
- Yuying Ma
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yi Wang
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Jing Xu
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhaobing Gao
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Bingqing Xia
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory for Immunological Research on Chronic Diseases, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
2
|
Zheng Q, Zhang H, Zhao H, Chen Y, Yang H, Li T, Cai Q, Chen Y, Wang Y, Zhang M, Zhang H. Ca 2+/calmodulin-dependent protein kinase II β decodes ER Ca 2+ transients to trigger autophagosome formation. Mol Cell 2025; 85:620-637.e6. [PMID: 39742665 DOI: 10.1016/j.molcel.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/16/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025]
Abstract
In multicellular organisms, very little is known about how Ca2+ transients on the ER outer surface elicited by autophagy stimuli are sustained and decoded to trigger autophagosome formation. Here, we show that Ca2+/calmodulin-dependent protein kinase II β (CaMKIIβ) integrates ER Ca2+ transients to trigger liquid-liquid phase separation (LLPS) of the autophagosome-initiating FIP200 complex. In response to ER Ca2+ transients, CaMKIIβ is recruited from actin filaments and forms condensates, which serve as sites for the emergence of or interaction with FIP200 puncta. CaMKIIβ phosphorylates FIP200 at Thr269, Thr1127, and Ser1484 to modulate LLPS and properties of the FIP200 complex, thereby controlling its function in autophagosome formation. CaMKIIβ also controls the amplitude, duration, and propagation of ER Ca2+ transients during autophagy induction. CaMKIIβ mutations identified in the neurodevelopmental disorder MRD54 affect the function of CaMKIIβ in autophagy. Our study reveals that CaMKIIβ is essential for sustaining and decoding ER Ca2+ transients to specify autophagosome formation in mammalian cells.
Collapse
Affiliation(s)
- Qiaoxia Zheng
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Huan Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Chen
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongzhining Yang
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Tingting Li
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Qixu Cai
- School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yingyu Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Youjun Wang
- College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Mingjie Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Pikor D, Hurła M, Słowikowski B, Szymanowicz O, Poszwa J, Banaszek N, Drelichowska A, Jagodziński PP, Kozubski W, Dorszewska J. Calcium Ions in the Physiology and Pathology of the Central Nervous System. Int J Mol Sci 2024; 25:13133. [PMID: 39684844 DOI: 10.3390/ijms252313133] [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: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Calcium ions play a key role in the physiological processes of the central nervous system. The intracellular calcium signal, in nerve cells, is part of the neurotransmission mechanism. They are responsible for stabilizing membrane potential and controlling the excitability of neurons. Calcium ions are a universal second messenger that participates in depolarizing signal transduction and contributes to synaptic activity. These ions take an active part in the mechanisms related to memory and learning. As a result of depolarization of the plasma membrane or stimulation of receptors, there is an extracellular influx of calcium ions into the cytosol or mobilization of these cations inside the cell, which increases the concentration of these ions in neurons. The influx of calcium ions into neurons occurs via plasma membrane receptors and voltage-dependent ion channels. Calcium channels play a key role in the functioning of the nervous system, regulating, among others, neuronal depolarization and neurotransmitter release. Channelopathies are groups of diseases resulting from mutations in genes encoding ion channel subunits, observed including the pathophysiology of neurological diseases such as migraine. A disturbed ability of neurons to maintain an appropriate level of calcium ions is also observed in such neurodegenerative processes as Alzheimer's disease, Parkinson's disease, Huntington's disease, and epilepsy. This review focuses on the involvement of calcium ions in physiological and pathological processes of the central nervous system. We also consider the use of calcium ions as a target for pharmacotherapy in the future.
Collapse
Affiliation(s)
- Damian Pikor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Joanna Poszwa
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Alicja Drelichowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
4
|
Yan G, Song R, Zhang J, Li Z, Lu Z, Liu Z, Zeng X, Yao J. MIF promotes Th17 cell differentiation in rheumatoid arthritis through ATF6 signal pathway. Mol Med 2024; 30:237. [PMID: 39614150 PMCID: PMC11605992 DOI: 10.1186/s10020-024-01005-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease that can lead to irreversible joint damage when it occurs, but its pathogenesis has not yet been elucidated. In this study, we explored the roles of macrophage migration inhibitory factor (MIF), endoplasmic reticulum stress (ER stress), and Th17 cells in the pathogenesis of RA. We have preliminarily confirmed that MIF expression in CD4+T cells and the proportion of Th17 cells are increased in active RA patients. We also found that ER stress is activated, initiating ATF6 pathway in the UPR. Additionally, using in vitro stimulation and co-immunoprecipitation experiments, we have confirmed the interaction between MIF and ATF6, which enhances protein expression in ATF6 pathway. Subsequently, in the chromatin immunoprecipitation assay, we observed the enrichment of ATF6 subunit on the promoter sequences of the Th17 cell differentiation genes STAT3 and RORC. Additionally, the differentiation of Th17 cells was disrupted by Ceapin-A7 (ATF6 inhibitor). In summary, our results indicate that MIF enhances ATF6 pathway signaling, which promotes the differentiation of Th17 cells. This could be a potential mechanism underlying the pathogenesis of RA, offering a new direction for the clinical treatment of RA.
Collapse
Affiliation(s)
- Guozhi Yan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, 528200, China
| | - Rongrong Song
- The Department of Laboratory Medicine, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde Foshan, Foshan, Guangdong, China
| | - Jieyu Zhang
- The Department of Laboratory Medicine, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde Foshan, Foshan, Guangdong, China
| | - Zhihao Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, 528200, China
| | - Zhantao Lu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, 528200, China
| | - Zijian Liu
- Department of Laboratory Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaokang Zeng
- Central Laboratory of The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, 528200, China.
| | - Jie Yao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, 528200, China.
- The Department of Laboratory Medicine, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde Foshan, Foshan, Guangdong, China.
| |
Collapse
|
5
|
Koszła O, Sołek P. Misfolding and aggregation in neurodegenerative diseases: protein quality control machinery as potential therapeutic clearance pathways. Cell Commun Signal 2024; 22:421. [PMID: 39215343 PMCID: PMC11365204 DOI: 10.1186/s12964-024-01791-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
The primary challenge in today's world of neuroscience is the search for new therapeutic possibilities for neurodegenerative disease. Central to these disorders lies among other factors, the aberrant folding, aggregation, and accumulation of proteins, resulting in the formation of toxic entities that contribute to neuronal degeneration. This review concentrates on the key proteins such as β-amyloid (Aβ), tau, and α-synuclein, elucidating the intricate molecular events underlying their misfolding and aggregation. We critically evaluate the molecular mechanisms governing the elimination of misfolded proteins, shedding light on potential therapeutic strategies. We specifically examine pathways such as the endoplasmic reticulum (ER) and unfolded protein response (UPR), chaperones, chaperone-mediated autophagy (CMA), and the intersecting signaling of Keap1-Nrf2-ARE, along with autophagy connected through p62. Above all, we emphasize the significance of these pathways as protein quality control mechanisms, encompassing interventions targeting protein aggregation, regulation of post-translational modifications, and enhancement of molecular chaperones and clearance. Additionally, we focus on current therapeutic possibilities and new, multi-target approaches. In conclusion, this review systematically consolidates insights into emerging therapeutic strategies predicated on protein aggregates clearance.
Collapse
Affiliation(s)
- Oliwia Koszła
- Department of Biopharmacy, Medical University of Lublin, 4A Chodzki St., Lublin, 20-093, Poland.
| | - Przemysław Sołek
- Department of Biopharmacy, Medical University of Lublin, 4A Chodzki St., Lublin, 20-093, Poland
- Department of Biochemistry and Toxicology, University of Life Sciences, 13 Akademicka St, Lublin, 20-950, Poland
| |
Collapse
|
6
|
Crapart CC, Scott ZC, Konno T, Sharma A, Parutto P, Bailey DMD, Westrate LM, Avezov E, Koslover EF. Luminal transport through intact endoplasmic reticulum limits the magnitude of localized Ca 2+ signals. Proc Natl Acad Sci U S A 2024; 121:e2312172121. [PMID: 38502705 PMCID: PMC10990089 DOI: 10.1073/pnas.2312172121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/09/2024] [Indexed: 03/21/2024] Open
Abstract
The endoplasmic reticulum (ER) forms an interconnected network of tubules stretching throughout the cell. Understanding how ER functionality relies on its structural organization is crucial for elucidating cellular vulnerability to ER perturbations, which have been implicated in several neuronal pathologies. One of the key functions of the ER is enabling Ca[Formula: see text] signaling by storing large quantities of this ion and releasing it into the cytoplasm in a spatiotemporally controlled manner. Through a combination of physical modeling and live-cell imaging, we demonstrate that alterations in ER shape significantly impact its ability to support efficient local Ca[Formula: see text] releases, due to hindered transport of luminal content within the ER. Our model reveals that rapid Ca[Formula: see text] release necessitates mobile luminal buffer proteins with moderate binding strength, moving through a well-connected network of ER tubules. These findings provide insight into the functional advantages of normal ER architecture, emphasizing its importance as a kinetically efficient intracellular Ca[Formula: see text] delivery system.
Collapse
Affiliation(s)
- Cécile C. Crapart
- UK Dementia Research Institute at the University of Cambridge, CambridgeCB2 0AH, United Kingdom
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, CambridgeCB2 0AH, United Kingdom
| | | | - Tasuku Konno
- UK Dementia Research Institute at the University of Cambridge, CambridgeCB2 0AH, United Kingdom
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, CambridgeCB2 0AH, United Kingdom
| | - Aman Sharma
- Department of Physics, University of California, San Diego, La Jolla, CA92130
| | - Pierre Parutto
- UK Dementia Research Institute at the University of Cambridge, CambridgeCB2 0AH, United Kingdom
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, CambridgeCB2 0AH, United Kingdom
| | - David M. D. Bailey
- UK Dementia Research Institute at the University of Cambridge, CambridgeCB2 0AH, United Kingdom
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, CambridgeCB2 0AH, United Kingdom
| | - Laura M. Westrate
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI49546
| | - Edward Avezov
- UK Dementia Research Institute at the University of Cambridge, CambridgeCB2 0AH, United Kingdom
- Department of Clinical Neurosciences, School of Clinical Medicine, The University of Cambridge, CambridgeCB2 0AH, United Kingdom
| | - Elena F. Koslover
- Department of Physics, University of California, San Diego, La Jolla, CA92130
| |
Collapse
|
7
|
Lin Z, Zhang X, Nandi P, Lin Y, Wang L, Chu YS, Paape T, Yang Y, Xiao X, Liu Q. Correlative single-cell hard X-ray computed tomography and X-ray fluorescence imaging. Commun Biol 2024; 7:280. [PMID: 38448784 PMCID: PMC10917812 DOI: 10.1038/s42003-024-05950-y] [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: 08/24/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
X-ray computed tomography (XCT) and X-ray fluorescence (XRF) imaging are two non-invasive imaging techniques to study cellular structures and chemical element distributions, respectively. However, correlative X-ray computed tomography and fluorescence imaging for the same cell have yet to be routinely realized due to challenges in sample preparation and X-ray radiation damage. Here we report an integrated experimental and computational workflow for achieving correlative multi-modality X-ray imaging of a single cell. The method consists of the preparation of radiation-resistant single-cell samples using live-cell imaging-assisted chemical fixation and freeze-drying procedures, targeting and labeling cells for correlative XCT and XRF measurement, and computational reconstruction of the correlative and multi-modality images. With XCT, cellular structures including the overall structure and intracellular organelles are visualized, while XRF imaging reveals the distribution of multiple chemical elements within the same cell. Our correlative method demonstrates the feasibility and broad applicability of using X-rays to understand cellular structures and the roles of chemical elements and related proteins in signaling and other biological processes.
Collapse
Affiliation(s)
- Zihan Lin
- Biology Department, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Xiao Zhang
- Biology Department, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Purbasha Nandi
- Biology Department, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Yuewei Lin
- Computational Science Initiative, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Liguo Wang
- Laboratory for BioMolecular Structure, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Yong S Chu
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, 11973, USA
| | - Timothy Paape
- Biology Department, Brookhaven National Laboratory, Upton, NY, 11973, USA
- U.S. Department of Agriculture's Agricultural Research Service at Children's Nutrition Research Center, Houston, TX, 77030, USA
| | - Yang Yang
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, 11973, USA.
| | - Xianghui Xiao
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, 11973, USA.
| | - Qun Liu
- Biology Department, Brookhaven National Laboratory, Upton, NY, 11973, USA.
- National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, 11973, USA.
| |
Collapse
|
8
|
Cao R, Cao C, Hu X, Du K, Zhang J, Li M, Li B, Lin H, Zhang A, Li Y, Wu L, Huang Y. Kaempferol attenuates carbon tetrachloride (CCl 4)-induced hepatic fibrosis by promoting ASIC1a degradation and suppression of the ASIC1a-mediated ERS. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155125. [PMID: 37820466 DOI: 10.1016/j.phymed.2023.155125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Kaempferol is a flavonoid derived from the herb, Kaempferia galanga L., in addition to exhibiting a wide range of pharmacological properties, kaempferol is also an anti-inflammatory, anti-lipid metabolizing, and anti-oxidative stress agent. The underlying molecular mechanisms of its effects on vascular endothelial growth factor (VEGF) secretion and activation of hepatic stellate cells (HSCs) are yet unknown. Activated HSCs induces VEGF release and extracellular matrix (ECM) accumulation which are important factors in hepatic fibrosis. PURPOSE Our aim is to explore how kaempferol may affect hepatic fibrosis and the mechanisms behind its effects. METHODS The in vivo model was Sprague-Dawley rats induced with carbon tetrachloride (CCl4). Histological staining was used to observe histological features of the liver. The levels of (alanine aminotransferase) ALT and (aspartate aminotransferase) AST were detected by the corresponding kits. Platelet-derived growth factor (PDGF) was used to stimulate the HSC-T6 rat hepatic stellate cells. The mechanisms underlying this process were investigated using a variety of molecular approaches, including immunofluorescence, RT-qPCR, and western blotting. Moreover, intracellular Ca2+ were observed by laser confocal microscope. RESULTS It was found that kaempferol significantly reduced the expression of ASIC1a, VEGF, α-SMA and Collagen-I proteins in a model of CCl4-induced hepatic fibrosis in rats. In HSC-T6, kaempferol inhibits activation of HSCs by decreasing expression of ASIC1a, eIF2α, p-eIF2α and ATF-4. Laser confocal fluorescence showed that kaempferol inhibited Ca2+ influx and reduced Ca2+ concentration around the endoplasmic reticulum. Molecular docking and cellular thermal shift assay (CETSA) results further indicated that kaempferol interacted with ASIC1a. We found that kaempferol may promote the degradation of ASIC1a and inhibited ASIC1a- mediated upregulation of ERS. CONCLUSION The data from our in vivo experiments demonstrate that kaempferol effectively attenuates hepatic fibrosis. In vitro studies we further propose a novel mechanism of kaempferol against hepatic fibrosis which can interact with ASIC1a and promote ASIC1a degradation while inhibiting the activation and VEGF release of HSCs by suppressing the ASIC1a-eIF2α-ATF-4 signaling pathway.
Collapse
Affiliation(s)
- Rui Cao
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Chun Cao
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Xiaojie Hu
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Kang Du
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Mengxue Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Bowen Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Huimin Lin
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Anqi Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Yangyang Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Li Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Yan Huang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
9
|
Hasan M, Al-Thani H, El-Menyar A, Zeidan A, Al-Thani A, Yalcin HC. Disturbed hemodynamics and oxidative stress interaction in endothelial dysfunction and AAA progression: Focus on Nrf2 pathway. Int J Cardiol 2023; 389:131238. [PMID: 37536420 DOI: 10.1016/j.ijcard.2023.131238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/30/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Hemodynamic shear stress is one of the major factors that are involved in the pathogenesis of many cardiovascular diseases including atherosclerosis and abdominal aortic aneurysm (AAA), through its modulatory effect on the endothelial cell's redox homeostasis and mechanosensitive gene expression. Among important mechanisms, oxidative stress, endoplasmic reticulum stress activation, and the subsequent endothelial dysfunction are attributed to disturbed blood flow and low shear stress in the vascular curvature and bifurcations which are considered atheroprone regions and aneurysm occurrence spots. Many pathways were shown to be involved in AAA progression. Of particular interest from recent findings is, the (Nrf2)/Keap-1 pathway, where Nrf2 is a transcription factor that has antioxidant properties and is strongly associated with several CVDs, yet, the exact mechanism by which Nrf2 alleviates CVDs still to be elucidated. Nrf2 expression is closely affected by shear stress and was shown to participate in AAA. In the current review paper, we discussed the link between disturbed hemodynamics and its effect on Nrf2 as a mechanosensitive gene and its role in the development of endothelial dysfunction which is linked to the progression of AAA.
Collapse
Affiliation(s)
- Maram Hasan
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ayman El-Menyar
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar; Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Asad Zeidan
- Department of Basic Sciences, College of Medicine, QU health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Asmaa Al-Thani
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
10
|
Zhang IX, Herrmann A, Leon J, Jeyarajan S, Arunagiri A, Arvan P, Gilon P, Satin LS. ER stress increases expression of intracellular calcium channel RyR1 to modify Ca 2+ homeostasis in pancreatic beta cells. J Biol Chem 2023; 299:105065. [PMID: 37468098 PMCID: PMC10448220 DOI: 10.1016/j.jbc.2023.105065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023] Open
Abstract
Pancreatic beta cells maintain glucose homeostasis by secreting pulses of insulin in response to a rise in plasma glucose. Pulsatile insulin secretion occurs as a result of glucose-induced oscillations in beta-cell cytosolic Ca2+. The endoplasmic reticulum (ER) helps regulate beta-cell cytosolic Ca2+, and ER stress can lead to ER Ca2+ reduction, beta-cell dysfunction, and an increased risk of type 2 diabetes. However, the mechanistic effects of ER stress on individual calcium channels are not well understood. To determine the effects of tunicamycin-induced ER stress on ER inositol 1,4,5-triphosphate receptors (IP3Rs) and ryanodine receptors (RyRs) and their involvement in subsequent Ca2+ dysregulation, we treated INS-1 832/13 cells and primary mouse islets with ER stress inducer tunicamycin (TM). We showed TM treatment increased RyR1 mRNA without affecting RyR2 mRNA and decreased both IP3R1 and IP3R3 mRNA. Furthermore, we found stress reduced ER Ca2+ levels, triggered oscillations in cytosolic Ca2+ under subthreshold glucose conditions, and increased apoptosis and that these changes were prevented by cotreatment with the RyR1 inhibitor dantrolene. In addition, we demonstrated silencing RyR1-suppressed TM-induced subthreshold cytosolic Ca2+ oscillations, but silencing RyR2 did not affect these oscillations. In contrast, inhibiting IP3Rs with xestospongin-C failed to suppress the TM-induced cytosolic Ca2+ oscillations and did not protect beta cells from TM-induced apoptosis although xestospongin-C inclusion did prevent ER Ca2+ reduction. Taken together, these results show changes in RyR1 play a critical role in ER stress-induced Ca2+ dysfunction and beta-cell apoptosis.
Collapse
Affiliation(s)
- Irina X Zhang
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Andrea Herrmann
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Juan Leon
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Sivakumar Jeyarajan
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Anoop Arunagiri
- Department of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter Arvan
- Department of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick Gilon
- Pole of Endocrinology, Diabetes and Nutrition (EDIN), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Leslie S Satin
- Department of Pharmacology and Brehm Diabetes Research Center, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| |
Collapse
|
11
|
Liu Y, Ma Y, Xu J, Zhang G, Zhao X, He Z, Wang L, Yin N, Peng M. VMP1 prevents Ca2+ overload in endoplasmic reticulum and maintains naive T cell survival. J Exp Med 2023; 220:e20221068. [PMID: 36971758 PMCID: PMC10060355 DOI: 10.1084/jem.20221068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/11/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
Ca2+ in endoplasmic reticulum (ER) dictates T cell activation, proliferation, and function via store-operated Ca2+ entry. How naive T cells maintain an appropriate level of Ca2+ in ER remains poorly understood. Here, we show that the ER transmembrane protein VMP1 is essential for maintaining ER Ca2+ homeostasis in naive T cells. VMP1 promotes Ca2+ release from ER under steady state, and its deficiency leads to ER Ca2+ overload, ER stress, and secondary Ca2+ overload in mitochondria, resulting in massive apoptosis of naive T cells and defective T cell response. Aspartic acid 272 (D272) of VMP1 is critical for its ER Ca2+ releasing activity, and a knockin mouse strain with D272 mutated to asparagine (D272N) demonstrates all functions of VMP1 in T cells in vivo depend on its regulation of ER Ca2+. These data uncover an indispensable role of VMP1 in preventing ER Ca2+ overload and maintaining naive T cell survival.
Collapse
Affiliation(s)
- Ying Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Yuying Ma
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Jing Xu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Guangyue Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xiaocui Zhao
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zihao He
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Lixia Wang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Min Peng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute for Immunology, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Furuta Y, Zhou Z. How do necrotic cells expose phosphatidylserine to attract their predators—What’s unique and what’s in common with apoptotic cells. Front Cell Dev Biol 2023; 11:1170551. [PMID: 37091984 PMCID: PMC10113483 DOI: 10.3389/fcell.2023.1170551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 04/09/2023] Open
Abstract
Phosphatidylserine (PS) is a lipid component of the plasma membrane. It is asymmetrically distributed to the inner leaflet in live cells. In cells undergoing apoptosis, phosphatidylserine is exposed to the outer surfaces. The exposed phosphatidylserine acts as an evolutionarily conserved “eat-me” signal that attracts neighboring engulfing cells in metazoan organisms, including the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and mammals. During apoptosis, the exposure of phosphatidylserine to the outer surface of a cell is driven by the membrane scramblases and flippases, the activities of which are regulated by caspases. Cells undergoing necrosis, a kind of cell death frequently associated with cellular injuries and morphologically distinct from apoptosis, were initially believed to allow passive exposure of phosphatidylserine through membrane rupture. Later studies revealed that necrotic cells actively expose phosphatidylserine before any rupture occurs. A recent study in C. elegans further reported that the calcium ion (Ca2+) plays an essential role in promoting the exposure of phosphatidylserine on the surfaces of necrotic cells. These findings indicate that necrotic and apoptotic cells, which die through different molecular mechanisms, use common and unique mechanisms for promoting the exposure of the same “eat me” signal. This article will review the mechanisms regulating the exposure of phosphatidylserine on the surfaces of necrotic and apoptotic cells and highlight their similarities and differences.
Collapse
|
13
|
Abstract
Inositol 1,4,5-trisphosphate (IP3) plays a key role in calcium signaling. After stimulation, it diffuses from the plasma membrane where it is produced to the endoplasmic reticulum where its receptors are localized. Based on in vitro measurements, IP3 was long thought to be a global messenger characterized by a diffusion coefficient of ~ 280 μm2s-1. However, in vivo observations revealed that this value does not match with the timing of localized Ca2+ increases induced by the confined release of a non-metabolizable IP3 analog. A theoretical analysis of these data concluded that in intact cells diffusion of IP3 is strongly hindered, leading to a 30-fold reduction of the diffusion coefficient. Here, we performed a new computational analysis of the same observations using a stochastic model of Ca2+ puffs. Our simulations concluded that the value of the effective IP3 diffusion coefficient is close to 100 μm2s-1. Such moderate reduction with respect to in vitro estimations quantitatively agrees with a buffering effect by non-fully bound inactive IP3 receptors. The model also reveals that IP3 spreading is not much affected by the endoplasmic reticulum, which represents an obstacle to the free displacement of molecules, but can be significantly increased in cells displaying elongated, 1-dimensional like geometries.
Collapse
|
14
|
Vestuto V, Di Sarno V, Musella S, Di Dona G, Moltedo O, Gomez-Monterrey IM, Bertamino A, Ostacolo C, Campiglia P, Ciaglia T. New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors? Int J Mol Sci 2022; 24:185. [PMID: 36613628 PMCID: PMC9820239 DOI: 10.3390/ijms24010185] [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: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic structure, playing multiple roles including calcium storage, protein synthesis and lipid metabolism. During cellular stress, variations in ER homeostasis and its functioning occur. This condition is referred as ER stress and generates a cascade of signaling events termed unfolded protein response (UPR), activated as adaptative response to mitigate the ER stress condition. In this regard, calcium levels play a pivotal role in ER homeostasis and therefore in cell fate regulation since calcium signaling is implicated in a plethora of physiological processes, but also in disease conditions such as neurodegeneration, cancer and metabolic disorders. A large body of emerging evidence highlighted the functional role of TRP channels and their ability to promote cell survival or death depending on endoplasmic reticulum stress resolution, making them an attractive target. Thus, in this review we focused on the TRP channels' correlation to UPR-mediated ER stress in disease pathogenesis, providing an overview of their implication in the activation of this cellular response.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Giorgio Di Dona
- Pineta Grande Hospital, Via Domiziana, km 30/00, 81030 Castel Volturno, CE, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | | | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- European Biomedical Research Institute of Salerno, Via S. De Renzi 50, 84125 Salerno, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| |
Collapse
|
15
|
Zhou X, Chen Z, Xiao L, Zhong Y, Liu Y, Wu J, Tao H. Intracellular calcium homeostasis and its dysregulation underlying epileptic seizures. Seizure 2022; 103:126-136. [DOI: 10.1016/j.seizure.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
|
16
|
Zheng Q, Chen Y, Chen D, Zhao H, Feng Y, Meng Q, Zhao Y, Zhang H. Calcium transients on the ER surface trigger liquid-liquid phase separation of FIP200 to specify autophagosome initiation sites. Cell 2022; 185:4082-4098.e22. [PMID: 36198318 DOI: 10.1016/j.cell.2022.09.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/20/2022] [Accepted: 08/30/2022] [Indexed: 02/06/2023]
Abstract
The mechanism that initiates autophagosome formation on the ER in multicellular organisms is elusive. Here, we showed that autophagy stimuli trigger Ca2+ transients on the outer surface of the ER membrane, whose amplitude, frequency, and duration are controlled by the metazoan-specific ER transmembrane autophagy protein EPG-4/EI24. Persistent Ca2+ transients/oscillations on the cytosolic ER surface in EI24-depleted cells cause accumulation of FIP200 autophagosome initiation complexes on the ER. This defect is suppressed by attenuating ER Ca2+ transients. Multi-modal SIM analysis revealed that Ca2+ transients on the ER trigger the formation of dynamic and fusion-prone liquid-like FIP200 puncta. Starvation-induced Ca2+ transients on lysosomes also induce FIP200 puncta that further move to the ER. Multiple FIP200 puncta on the ER, whose association depends on the ER proteins VAPA/B and ATL2/3, assemble into autophagosome formation sites. Thus, Ca2+ transients are crucial for triggering phase separation of FIP200 to specify autophagosome initiation sites in metazoans.
Collapse
Affiliation(s)
- Qiaoxia Zheng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Di Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yun Feng
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Quan Meng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yan Zhao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
17
|
Parys JB, Van Coppenolle F. Sec61 complex/translocon: The role of an atypical ER Ca 2+-leak channel in health and disease. Front Physiol 2022; 13:991149. [PMID: 36277220 PMCID: PMC9582130 DOI: 10.3389/fphys.2022.991149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2023] Open
Abstract
The heterotrimeric Sec61 protein complex forms the functional core of the so-called translocon that forms an aqueous channel in the endoplasmic reticulum (ER). The primary role of the Sec61 complex is to allow protein import in the ER during translation. Surprisingly, a completely different function in intracellular Ca2+ homeostasis has emerged for the Sec61 complex, and the latter is now accepted as one of the major Ca2+-leak pathways of the ER. In this review, we first discuss the structure of the Sec61 complex and focus on the pharmacology and regulation of the Sec61 complex as a Ca2+-leak channel. Subsequently, we will pay particular attention to pathologies that are linked to Sec61 mutations, such as plasma cell deficiency and congenital neutropenia. Finally, we will explore the relevance of the Sec61 complex as a Ca2+-leak channel in various pathophysiological (ER stress, apoptosis, ischemia-reperfusion) and pathological (type 2 diabetes, cancer) settings.
Collapse
Affiliation(s)
- Jan B. Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Fabien Van Coppenolle
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Groupement Hospitalier EST, Department of Cardiology, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
18
|
Deficiency of the Two-Pore Potassium Channel KCNK9 Impairs Intestinal Epithelial Cell Survival and Aggravates Dextran Sodium Sulfate-Induced Colitis. Cell Mol Gastroenterol Hepatol 2022; 14:1199-1211. [PMID: 35973573 PMCID: PMC9579309 DOI: 10.1016/j.jcmgh.2022.08.003] [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: 05/03/2021] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS The 2-pore potassium channel subfamily K member 9 (KCNK9) regulates intracellular calcium concentration and thus modulates cell survival and inflammatory signaling pathways. It also was recognized as a risk allele for inflammatory bowel disease. However, it remains unclear whether KCNK9 modulates inflammatory bowel disease via its impact on immune cell function or whether its influence on calcium homeostasis also is relevant in intestinal epithelial cells. METHODS Kcnk9-/- mice were challenged with 3% dextran sulfate sodium (DSS) to induce experimental acute colitis. Primary cultures of intestinal epithelial cells were generated, and expression of potassium channels as well as cytosolic calcium levels and susceptibility to apoptosis were evaluated. Furthermore, we evaluated whether KCNK9 deficiency was compensated by the closely related 2-pore potassium channel KCNK3 in vivo or in vitro. RESULTS Compared with controls, KCNK9 deficiency or its pharmacologic blockade were associated with aggravated DSS-induced colitis compared with wild-type animals. In the absence of KCNK9, intestinal epithelial cells showed increased intracellular calcium levels and were more prone to mitochondrial damage and caspase-9-dependent apoptosis. We found that expression of KCNK3 was increased in Kcnk9-/- mice but did not prevent apoptosis after DSS exposure. Conversely, increased levels of KCNK9 in Kcnk3-/- mice were associated with an ameliorated course of DSS-induced colitis. CONCLUSIONS KCNK9 enhances mitochondrial stability, reduces apoptosis, und thus supports epithelial cell survival after DSS exposure in vivo and in vitro. Conversely, its increased expression in Kcnk3-/- resulted in less mitochondrial damage and apoptosis and was associated with beneficial outcomes in DSS-induced colitis.
Collapse
|
19
|
Yang KY, Zhao S, Feng H, Shen J, Chen Y, Wang ST, Wang SJ, Zhang YX, Wang Y, Guo C, Liu H, Tang TS. Ca 2+ homeostasis maintained by TMCO1 underlies corpus callosum development via ERK signaling. Cell Death Dis 2022; 13:674. [PMID: 35927240 PMCID: PMC9352667 DOI: 10.1038/s41419-022-05131-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 01/21/2023]
Abstract
Transmembrane of coiled-coil domains 1 (TMCO1) plays an important role in maintaining homeostasis of calcium (Ca2+) stores in the endoplasmic reticulum (ER). TMCO1-defect syndrome shares multiple features with human cerebro-facio-thoracic (CFT) dysplasia, including abnormal corpus callosum (CC). Here, we report that TMCO1 is required for the normal development of CC through sustaining Ca2+ homeostasis. Tmco1-/- mice exhibit severe agenesis of CC with stalled white matter fiber bundles failing to pass across the midline. Mechanistically, the excessive Ca2+ signals caused by TMCO1 deficiency result in upregulation of FGFs and over-activation of ERK, leading to an excess of glial cell migration and overpopulated midline glia cells in the indusium griseum which secretes Slit2 to repulse extension of the neural fiber bundles before crossing the midline. Supportingly, using the clinical MEK inhibitors to attenuate the over-activated FGF/ERK signaling can significantly improve the CC formation in Tmco1-/- brains. Our findings not only unravel the underlying mechanism of abnormal CC in TMCO1 defect syndrome, but also offer an attractive prevention strategy to relieve the related agenesis of CC in patients.
Collapse
Affiliation(s)
- Ke-Yan Yang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Song Zhao
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Haiping Feng
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Jiaqi Shen
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yuwei Chen
- grid.410726.60000 0004 1797 8419Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101 China
| | - Si-Tong Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Si-Jia Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yu-Xin Zhang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yun Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Caixia Guo
- grid.410726.60000 0004 1797 8419Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101 China
| | - Hongmei Liu
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China ,grid.9227.e0000000119573309Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China ,grid.512959.3Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| | - Tie-Shan Tang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China ,grid.9227.e0000000119573309Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101 China ,grid.512959.3Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| |
Collapse
|
20
|
Spolaor S, Rovetta M, Nobile MS, Cazzaniga P, Tisi R, Besozzi D. Modeling Calcium Signaling in S. cerevisiae Highlights the Role and Regulation of the Calmodulin-Calcineurin Pathway in Response to Hypotonic Shock. Front Mol Biosci 2022; 9:856030. [PMID: 35664674 PMCID: PMC9158465 DOI: 10.3389/fmolb.2022.856030] [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: 01/16/2022] [Accepted: 04/04/2022] [Indexed: 01/17/2023] Open
Abstract
Calcium homeostasis and signaling processes in Saccharomyces cerevisiae, as well as in any eukaryotic organism, depend on various transporters and channels located on both the plasma and intracellular membranes. The activity of these proteins is regulated by a number of feedback mechanisms that act through the calmodulin-calcineurin pathway. When exposed to hypotonic shock (HTS), yeast cells respond with an increased cytosolic calcium transient, which seems to be conditioned by the opening of stretch-activated channels. To better understand the role of each channel and transporter involved in the generation and recovery of the calcium transient—and of their feedback regulations—we defined and analyzed a mathematical model of the calcium signaling response to HTS in yeast cells. The model was validated by comparing the simulation outcomes with calcium concentration variations before and during the HTS response, which were observed experimentally in both wild-type and mutant strains. Our results show that calcium normally enters the cell through the High Affinity Calcium influx System and mechanosensitive channels. The increase of the plasma membrane tension, caused by HTS, boosts the opening probability of mechanosensitive channels. This event causes a sudden calcium pulse that is rapidly dissipated by the activity of the vacuolar transporter Pmc1. According to model simulations, the role of another vacuolar transporter, Vcx1, is instead marginal, unless calcineurin is inhibited or removed. Our results also suggest that the mechanosensitive channels are subject to a calcium-dependent feedback inhibition, possibly involving calmodulin. Noteworthy, the model predictions are in accordance with literature results concerning some aspects of calcium homeostasis and signaling that were not specifically addressed within the model itself, suggesting that it actually depicts all the main cellular components and interactions that constitute the HTS calcium pathway, and thus can correctly reproduce the shaping of the calcium signature by calmodulin- and calcineurin-dependent complex regulations. The model predictions also allowed to provide an interpretation of different regulatory schemes involved in calcium handling in both wild-type and mutants yeast strains. The model could be easily extended to represent different calcium signals in other eukaryotic cells.
Collapse
Affiliation(s)
- Simone Spolaor
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Mattia Rovetta
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Marco S. Nobile
- Department of Environmental Sciences, Informatics and Statistics, Ca’ Foscari University of Venice, Venice, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
| | - Paolo Cazzaniga
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
- Department of Human and Social Sciences, University of Bergamo, Bergamo, Italy
| | - Renata Tisi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- *Correspondence: Renata Tisi, ; Daniela Besozzi,
| | - Daniela Besozzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre—B4, Milan, Italy
- SYSBIO/ISBE.IT Centre of Systems Biology, Milan, Italy
- *Correspondence: Renata Tisi, ; Daniela Besozzi,
| |
Collapse
|
21
|
Tirincsi A, Sicking M, Hadzibeganovic D, Haßdenteufel S, Lang S. The Molecular Biodiversity of Protein Targeting and Protein Transport Related to the Endoplasmic Reticulum. Int J Mol Sci 2021; 23:143. [PMID: 35008565 PMCID: PMC8745461 DOI: 10.3390/ijms23010143] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Looking at the variety of the thousands of different polypeptides that have been focused on in the research on the endoplasmic reticulum from the last five decades taught us one humble lesson: no one size fits all. Cells use an impressive array of components to enable the safe transport of protein cargo from the cytosolic ribosomes to the endoplasmic reticulum. Safety during the transit is warranted by the interplay of cytosolic chaperones, membrane receptors, and protein translocases that together form functional networks and serve as protein targeting and translocation routes. While two targeting routes to the endoplasmic reticulum, SRP (signal recognition particle) and GET (guided entry of tail-anchored proteins), prefer targeting determinants at the N- and C-terminus of the cargo polypeptide, respectively, the recently discovered SND (SRP-independent) route seems to preferentially cater for cargos with non-generic targeting signals that are less hydrophobic or more distant from the termini. With an emphasis on targeting routes and protein translocases, we will discuss those functional networks that drive efficient protein topogenesis and shed light on their redundant and dynamic nature in health and disease.
Collapse
Affiliation(s)
- Andrea Tirincsi
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany; (A.T.); (M.S.); (D.H.)
| | - Mark Sicking
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany; (A.T.); (M.S.); (D.H.)
| | - Drazena Hadzibeganovic
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany; (A.T.); (M.S.); (D.H.)
| | - Sarah Haßdenteufel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sven Lang
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany; (A.T.); (M.S.); (D.H.)
| |
Collapse
|
22
|
Yang F, Ma H, Butler MR, Ding XQ. Preservation of endoplasmic reticulum (ER) Ca 2+ stores by deletion of inositol-1,4,5-trisphosphate receptor type 1 promotes ER retrotranslocation, proteostasis, and protein outer segment localization in cyclic nucleotide-gated channel-deficient cone photoreceptors. FASEB J 2021; 35:e21579. [PMID: 33960001 DOI: 10.1096/fj.202002711r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/03/2021] [Accepted: 03/25/2021] [Indexed: 11/11/2022]
Abstract
Endoplasmic reticulum (ER) Ca2+ homeostasis relies on an appropriate balance between efflux- and influx-channel activity responding to dynamic changes of intracellular Ca2+ levels. Dysregulation of this complex signaling network has been shown to contribute to neuronal and photoreceptor death in neuro- and retinal degenerative diseases, respectively. In mice with cone cyclic nucleotide-gated (CNG) channel deficiency, a model of achromatopsia/cone dystrophy, cones display early-onset ER stress-associated apoptosis and protein mislocalization. Cones in these mice also show reduced cytosolic Ca2+ level and subsequent elevation in the ER Ca2+ -efflux-channel activity, specifically the inositol-1,4,5-trisphosphate receptor type 1 (IP3 R1), and deletion of IP3 R1 results in preservation of cones. This work investigated how preservation of ER Ca2+ stores leads to cone protection. We examined the effects of cone specific deletion of IP3 R1 on ER stress responses/cone death, protein localization, and ER proteostasis/ER-associated degradation. We demonstrated that deletion of IP3 R1 improves trafficking of cone-specific proteins M-/S-opsin and phosphodiesterase 6C to cone outer segments and reduces localization to cone inner segments. Consistent with the improved protein localization, deletion of IP3 R1 results in increased ER retrotranslocation protein expression, reduced proteasome subunit expression, reduced ER stress/cone death, and reduced retinal remodeling. We also observed the enhanced ER retrotranslocation in mice that have been treated with a chemical chaperone, supporting the connection between improved ER retrotranslocation/proteostasis and alleviation of ER stress. Findings from this work demonstrate the importance of ER Ca2+ stores in ER proteostasis and protein trafficking/localization in photoreceptors, strengthen the link between dysregulation of ER Ca2+ homeostasis and ER stress/cone degeneration, and support an involvement of improved ER proteostasis in ER Ca2+ preservation-induced cone protection; thereby identifying IP3 R1 as a critical mediator of ER stress and protein mislocalization and as a potential target to preserve cones in CNG channel deficiency.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hongwei Ma
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael R Butler
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
23
|
Jansen S, Baulain U, Habig C, Ramzan F, Schauer J, Schmitt AO, Scholz AM, Sharifi AR, Weigend A, Weigend S. Identification and Functional Annotation of Genes Related to Bone Stability in Laying Hens Using Random Forests. Genes (Basel) 2021; 12:702. [PMID: 34066823 PMCID: PMC8151682 DOI: 10.3390/genes12050702] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal disorders, including fractures and osteoporosis, in laying hens cause major welfare and economic problems. Although genetics have been shown to play a key role in bone integrity, little is yet known about the underlying genetic architecture of the traits. This study aimed to identify genes associated with bone breaking strength and bone mineral density of the tibiotarsus and the humerus in laying hens. Potentially informative single nucleotide polymorphisms (SNP) were identified using Random Forests classification. We then searched for genes known to be related to bone stability in close proximity to the SNPs and identified 16 potential candidates. Some of them had human orthologues. Based on our findings, we can support the assumption that multiple genes determine bone strength, with each of them having a rather small effect, as illustrated by our SNP effect estimates. Furthermore, the enrichment analysis showed that some of these candidates are involved in metabolic pathways critical for bone integrity. In conclusion, the identified candidates represent genes that may play a role in the bone integrity of chickens. Although further studies are needed to determine causality, the genes reported here are promising in terms of alleviating bone disorders in laying hens.
Collapse
Affiliation(s)
- Simon Jansen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
| | - Ulrich Baulain
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
| | - Christin Habig
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
| | - Faisal Ramzan
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, 37075 Göttingen, Germany; (F.R.); (A.O.S.)
| | - Jens Schauer
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
| | - Armin Otto Schmitt
- Breeding Informatics Group, Department of Animal Sciences, University of Göttingen, 37075 Göttingen, Germany; (F.R.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), University of Göttingen, 37075 Göttingen, Germany;
| | - Armin Manfred Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 85764 Oberschleissheim, Germany;
| | - Ahmad Reza Sharifi
- Center for Integrated Breeding Research (CiBreed), University of Göttingen, 37075 Göttingen, Germany;
- Animal Breeding and Genetics Group, Department of Animal Sciences, University of Göttingen, 37075 Göttingen, Germany
| | - Annett Weigend
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
| | - Steffen Weigend
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535 Neustadt, Germany; (U.B.); (C.H.); (J.S.); (A.W.); (S.W.)
- Center for Integrated Breeding Research (CiBreed), University of Göttingen, 37075 Göttingen, Germany;
| |
Collapse
|
24
|
Sicking M, Lang S, Bochen F, Roos A, Drenth JPH, Zakaria M, Zimmermann R, Linxweiler M. Complexity and Specificity of Sec61-Channelopathies: Human Diseases Affecting Gating of the Sec61 Complex. Cells 2021; 10:1036. [PMID: 33925740 PMCID: PMC8147068 DOI: 10.3390/cells10051036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
The rough endoplasmic reticulum (ER) of nucleated human cells has crucial functions in protein biogenesis, calcium (Ca2+) homeostasis, and signal transduction. Among the roughly one hundred components, which are involved in protein import and protein folding or assembly, two components stand out: The Sec61 complex and BiP. The Sec61 complex in the ER membrane represents the major entry point for precursor polypeptides into the membrane or lumen of the ER and provides a conduit for Ca2+ ions from the ER lumen to the cytosol. The second component, the Hsp70-type molecular chaperone immunoglobulin heavy chain binding protein, short BiP, plays central roles in protein folding and assembly (hence its name), protein import, cellular Ca2+ homeostasis, and various intracellular signal transduction pathways. For the purpose of this review, we focus on these two components, their relevant allosteric effectors and on the question of how their respective functional cycles are linked in order to reconcile the apparently contradictory features of the ER membrane, selective permeability for precursor polypeptides, and impermeability for Ca2+. The key issues are that the Sec61 complex exists in two conformations: An open and a closed state that are in a dynamic equilibrium with each other, and that BiP contributes to its gating in both directions in cooperation with different co-chaperones. While the open Sec61 complex forms an aqueous polypeptide-conducting- and transiently Ca2+-permeable channel, the closed complex is impermeable even to Ca2+. Therefore, we discuss the human hereditary and tumor diseases that are linked to Sec61 channel gating, termed Sec61-channelopathies, as disturbances of selective polypeptide-impermeability and/or aberrant Ca2+-permeability.
Collapse
Affiliation(s)
- Mark Sicking
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Sven Lang
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Florian Bochen
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| | - Andreas Roos
- Department of Neuropediatrics, Essen University Hospital, D-45147 Essen, Germany;
| | - Joost P. H. Drenth
- Department of Molecular Gastroenterology and Hepatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Muhammad Zakaria
- Department of Genetics, Hazara University, Mansehra 21300, Pakistan;
| | - Richard Zimmermann
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| |
Collapse
|
25
|
Mozolewski P, Jeziorek M, Schuster CM, Bading H, Frost B, Dobrowolski R. The role of nuclear Ca2+ in maintaining neuronal homeostasis and brain health. J Cell Sci 2021; 134:jcs254904. [PMID: 33912918 PMCID: PMC8084578 DOI: 10.1242/jcs.254904] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nuclear Ca2+ has emerged as one of the most potent mediators of the dialogue between neuronal synapses and the nucleus that regulates heterochromatin states, transcription factor activity, nuclear morphology and neuronal gene expression induced by synaptic activity. Recent studies underline the importance of nuclear Ca2+ signaling in long-lasting, activity-induced adaptation and maintenance of proper brain function. Diverse forms of neuroadaptation require transient nuclear Ca2+ signaling and cyclic AMP-responsive element-binding protein (CREB1, referred to here as CREB) as its prime target, which works as a tunable switch to drive and modulate specific gene expression profiles associated with memory, pain, addiction and neuroprotection. Furthermore, a reduction of nuclear Ca2+ levels has been shown to be neurotoxic and a causal factor driving the progression of neurodegenerative disorders, as well as affecting neuronal autophagy. Because of its central role in the brain, deficits in nuclear Ca2+ signaling may underlie a continuous loss of neuroprotection in the aging brain, contributing to the pathophysiology of Alzheimer's disease. In this Review, we discuss the principles of the 'nuclear calcium hypothesis' in the context of human brain function and its role in controlling diverse forms of neuroadaptation and neuroprotection. Furthermore, we present the most relevant and promising perspectives for future studies.
Collapse
Affiliation(s)
- Pawel Mozolewski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Maciej Jeziorek
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Christoph M. Schuster
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 345 and INF 366, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 345 and INF 366, 69120 Heidelberg, Germany
| | - Bess Frost
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health, San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
26
|
Lemos FO, Bultynck G, Parys JB. A comprehensive overview of the complex world of the endo- and sarcoplasmic reticulum Ca 2+-leak channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119020. [PMID: 33798602 DOI: 10.1016/j.bbamcr.2021.119020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Inside cells, the endoplasmic reticulum (ER) forms the largest Ca2+ store. Ca2+ is actively pumped by the SERCA pumps in the ER, where intraluminal Ca2+-binding proteins enable the accumulation of large amount of Ca2+. IP3 receptors and the ryanodine receptors mediate the release of Ca2+ in a controlled way, thereby evoking complex spatio-temporal signals in the cell. The steady state Ca2+ concentration in the ER of about 500 μM results from the balance between SERCA-mediated Ca2+ uptake and the passive leakage of Ca2+. The passive Ca2+ leak from the ER is often ignored, but can play an important physiological role, depending on the cellular context. Moreover, excessive Ca2+ leakage significantly lowers the amount of Ca2+ stored in the ER compared to normal conditions, thereby limiting the possibility to evoke Ca2+ signals and/or causing ER stress, leading to pathological consequences. The so-called Ca2+-leak channels responsible for Ca2+ leakage from the ER are however still not well understood, despite over 20 different proteins have been proposed to contribute to it. This review has the aim to critically evaluate the available evidence about the various channels potentially involved and to draw conclusions about their relative importance.
Collapse
Affiliation(s)
- Fernanda O Lemos
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
27
|
Chen TT, Zhou X, Xu YN, Li Y, Wu XY, Xiang Q, Fu LY, Hu XX, Tao L, Shen XC. Gastrodin ameliorates learning and memory impairment in rats with vascular dementia by promoting autophagy flux via inhibition of the Ca 2+/CaMKII signal pathway. Aging (Albany NY) 2021; 13:9542-9565. [PMID: 33714957 PMCID: PMC8064221 DOI: 10.18632/aging.202667] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
Vascular dementia (VD) is a common disease that occurs during human aging. Gastrodin (GAS) has potential benefits for the prevention and treatment of VD. In the present study, we investigated the effects of GAS on cognitive dysfunction in rats with VD induced by permanent middle cerebral artery occlusion (pMCAO) and explored the underlying mechanism. Immunohistochemical and western blot analyses revealed that GAS attenuated hippocampal levels of LC3 (microtubule-associated protein 1 light chain 3), p62, and phosphorylated CaMKII (Ca2+-calmodulin stimulated protein kinase II) in VD rats. Additionally, our results revealed that cobalt chloride blocked autophagic flux in HT22 cells, which was confirmed by increased levels of LC3 and p62 when combined with chloroquine. Notably, GAS ameliorated the impaired autophagic flux. Furthermore, we confirmed that GAS combined with KN93 (a CaMKII inhibitor) or CaMKII knockdown did not impact the reduced p62 levels when compared with GAS treatment alone. Furthermore, a co-immunoprecipitation assay demonstrated that endogenous p62 bound to CaMKII, as confirmed by mass spectrometric analysis after the immunoprecipitation of p62 from HT22 cells. These findings revealed that GAS attenuated autophagic flux dysfunction by inhibiting the Ca2+/CaMKII signaling pathway to ameliorate cognitive impairment in VD.
Collapse
Affiliation(s)
- Ting-Ting Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,Guiyang Maternal and Child Health-Care Hospital, Guiyang 550000, P.R. China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources and The Union Key Laboratory of Guiyang City, Guizhou Medical University, School of Pharmaceutical Sciences, Guiyang 550025, P.R. China
| | - Xue Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Yi-Ni Xu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Yue Li
- Guiyang Maternal and Child Health-Care Hospital, Guiyang 550000, P.R. China
| | - Xiao-Ying Wu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources and The Union Key Laboratory of Guiyang City, Guizhou Medical University, School of Pharmaceutical Sciences, Guiyang 550025, P.R. China
| | - Quan Xiang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Ling-Yun Fu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Xiao-Xia Hu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Ling Tao
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Xiang-Chun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, P.R. China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources and The Union Key Laboratory of Guiyang City, Guizhou Medical University, School of Pharmaceutical Sciences, Guiyang 550025, P.R. China.,The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| |
Collapse
|
28
|
Furuta Y, Pena-Ramos O, Li Z, Chiao L, Zhou Z. Calcium ions trigger the exposure of phosphatidylserine on the surface of necrotic cells. PLoS Genet 2021; 17:e1009066. [PMID: 33571185 PMCID: PMC7904182 DOI: 10.1371/journal.pgen.1009066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/24/2021] [Accepted: 01/18/2021] [Indexed: 11/18/2022] Open
Abstract
Intracellular Ca2+ level is under strict regulation through calcium channels and storage pools including the endoplasmic reticulum (ER). Mutations in certain ion channel subunits, which cause mis-regulated Ca2+ influx, induce the excitotoxic necrosis of neurons. In the nematode Caenorhabditis elegans, dominant mutations in the DEG/ENaC sodium channel subunit MEC-4 induce six mechanosensory (touch) neurons to undergo excitotoxic necrosis. These necrotic neurons are subsequently engulfed and digested by neighboring hypodermal cells. We previously reported that necrotic touch neurons actively expose phosphatidylserine (PS), an “eat-me” signal, to attract engulfing cells. However, the upstream signal that triggers PS externalization remained elusive. Here we report that a robust and transient increase of cytoplasmic Ca2+ level occurs prior to the exposure of PS on necrotic touch neurons. Inhibiting the release of Ca2+ from the ER, either pharmacologically or genetically, specifically impairs PS exposure on necrotic but not apoptotic cells. On the contrary, inhibiting the reuptake of cytoplasmic Ca2+ into the ER induces ectopic necrosis and PS exposure. Remarkably, PS exposure occurs independently of other necrosis events. Furthermore, unlike in mutants of DEG/ENaC channels, in dominant mutants of deg-3 and trp-4, which encode Ca2+ channels, PS exposure on necrotic neurons does not rely on the ER Ca2+ pool. Our findings indicate that high levels of cytoplasmic Ca2+ are necessary and sufficient for PS exposure. They further reveal two Ca2+-dependent, necrosis-specific pathways that promote PS exposure, a “two-step” pathway initiated by a modest influx of Ca2+ and further boosted by the release of Ca2+ from the ER, and another, ER-independent, pathway. Moreover, we found that ANOH-1, the worm homolog of mammalian phospholipid scramblase TMEM16F, is necessary for efficient PS exposure in thapsgargin-treated worms and trp-4 mutants, like in mec-4 mutants. We propose that both the ER-mediated and ER-independent Ca2+ pathways promote PS externalization through activating ANOH-1. Necrosis is a type of cell death that exhibits distinct morphological features such as cell and organelle swelling. Necrotic cells expose phosphatidylserine (PS)–a type of phospholipid—on their outer surfaces. Receptor molecules on phagocytes detect PS on necrotic cells and subsequently initiate the engulfment process. As necrosis is associated with stroke, cancer, neurodegenerative diseases, and heart diseases, studying necrotic cell clearance has important medical relevance. In the model organism the nematode C. elegans, we previously identified membrane proteins that promote the exposure of PS on necrotic cell surfaces by studying neurons that are induced to undergo necrosis by dominant mutations in ion channels. Here, in C. elegans, we have discovered that the necrotic insults trigger an increase of the cytoplasmic calcium ion (Ca2+), which in turn promotes PS externalization on necrotic cell surfaces. Furthermore, we have identified two different mechanisms that increase cytoplasmic Ca2+ levels, one dependent on the Ca2+ contribution from the endoplasmic reticulum (ER), the other independent of the ER. The Ca2+ signal targets ANOH-1, a worm homolog of mammalian proteins capable of externalizing PS, for promoting PS exposure on necrotic cells. Our findings reveal novel upstream regulatory mechanisms that promote necrotic cell clearance in animals.
Collapse
Affiliation(s)
- Yoshitaka Furuta
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- School of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, Japan
| | - Omar Pena-Ramos
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zao Li
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lucia Chiao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zheng Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
29
|
Ismatullah H, Jabeen I, Saeed MT. Biological Regulatory Network (BRN) Analysis and Molecular Docking Simulations to Probe the Modulation of IP 3R Mediated Ca 2+ Signaling in Cancer. Genes (Basel) 2020; 12:34. [PMID: 33383780 PMCID: PMC7823498 DOI: 10.3390/genes12010034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Inositol trisphosphate receptor (IP3R) mediated Ca+2 signaling is essential in determining the cell fate by regulating numerous cellular processes, including cell division and cell death. Despite extensive studies about the characterization of IP3R in cancer, the underlying molecular mechanism initiating the cell proliferation and apoptosis remained enigmatic. Moreover, in cancer, the modulation of IP3R in downstream signaling pathways, which control oncogenesis and cancer progression, is not well characterized. Here, we constructed a biological regulatory network (BRN), and describe the remodeling of IP3R mediated Ca2+ signaling as a central key that controls the cellular processes in cancer. Moreover, we summarize how the inhibition of IP3R affects the deregulated cell proliferation and cell death in cancer cells and results in the initiation of pro-survival responses in resistance of cell death in normal cells. Further, we also investigated the role of stereo-specificity of IP3 molecule and its analogs in binding with the IP3 receptor. Molecular docking simulations showed that the hydroxyl group at R6 position along with the phosphate group at R5 position in 'R' conformation is more favorable for IP3 interactions. Additionally, Arg-266 and Arg-510 showed π-π and hydrogen bond interactions and Ser-278 forms hydrogen bond interactions with the IP3 binding site. Thus, they are identified as crucial for the binding of antagonists.
Collapse
Affiliation(s)
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Academic-I Building, H-12 Islamabad 44000, Pakistan; (H.I.); (M.T.S.)
| | | |
Collapse
|
30
|
Cremer T, Neefjes J, Berlin I. The journey of Ca 2+ through the cell - pulsing through the network of ER membrane contact sites. J Cell Sci 2020; 133:133/24/jcs249136. [PMID: 33376155 DOI: 10.1242/jcs.249136] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcium is the third most abundant metal on earth, and the fundaments of its homeostasis date back to pre-eukaryotic life forms. In higher organisms, Ca2+ serves as a cofactor for a wide array of (enzymatic) interactions in diverse cellular contexts and constitutes the most important signaling entity in excitable cells. To enable responsive behavior, cytosolic Ca2+ concentrations are kept low through sequestration into organellar stores, particularly the endoplasmic reticulum (ER), but also mitochondria and lysosomes. Specific triggers are then used to instigate a local release of Ca2+ on demand. Here, communication between organelles comes into play, which is accomplished through intimate yet dynamic contacts, termed membrane contact sites (MCSs). The field of MCS biology in relation to cellular Ca2+ homeostasis has exploded in recent years. Taking advantage of this new wealth of knowledge, in this Review, we invite the reader on a journey of Ca2+ flux through the ER and its associated MCSs. New mechanistic insights and technological advances inform the narrative on Ca2+ acquisition and mobilization at these sites of communication between organelles, and guide the discussion of their consequences for cellular physiology.
Collapse
Affiliation(s)
- Tom Cremer
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Ilana Berlin
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| |
Collapse
|
31
|
Hu H, Jiang M, Cao Y, Zhang Z, Jiang B, Tian F, Feng J, Dou Y, Gorospe M, Zheng M, Zheng L, Yang Z, Wang W. HuR regulates phospholamban expression in isoproterenol-induced cardiac remodelling. Cardiovasc Res 2020; 116:944-955. [PMID: 31373621 DOI: 10.1093/cvr/cvz205] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/30/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022] Open
Abstract
AIMS The elevated expression of phospholamban (PLB) has been observed in heart failure and cardiac remodelling, inhibiting the affinity of Ca2+ pump to Ca2+ thereby impairing heart relaxation. However, the mechanisms underlying the regulation of PLB remains to be further studied. The present study aims to test the role of RNA-binding protein HuR in the regulation of PLB and the impact of this regulatory process in cardiac remodelling. METHODS AND RESULTS A mouse model specifically deleted HuR in cardiomyocytes were used for testing the role of HuR in regulating PLB during isoproterenol (ISO)-induced cardiac remodelling. HuR deficiency did not significantly influence the phenotype and function of mouse heart under static status. However, deletion of HuR in cardiomyocytes mitigated the effect of ISO in inducing PLB expression and reducing β1-AR expression, in turn aggravating ISO-induced myocardial hypertrophy and cardiac fibrosis. In H9C2 cells, association of HuR with PLB and β1-AR mRNAs stabilized PLB mRNA and destabilized β1-AR mRNA, respectively. CONCLUSION HuR stabilizes PLB mRNA and destabilizes β1-AR mRNA. The HuR-PLB and HuR-β1-AR regulatory processes impact on ISO-induced cardiac remodelling.
Collapse
Affiliation(s)
- Han Hu
- >Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Mingyang Jiang
- Department of Cardiology, State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Yangpo Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road Beijing, 100191, China
| | - Zhuojun Zhang
- >Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Bin Jiang
- >Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Feng Tian
- Department of Laboratory Animal Science, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Juan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road Beijing, 100191, China
| | - Yali Dou
- Department of Pathology and Biological Chemistry, University of Michigan, 1301 Catherine Street, Ann Arbor, MI 48105, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Ming Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road Beijing, 100191, China
| | - Lemin Zheng
- Institute of Cardiovascular Research, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| | - Zhongzhou Yang
- Department of Cardiology, State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210061, China
| | - Wengong Wang
- >Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, China
| |
Collapse
|
32
|
Al-Mawla R, Ducrozet M, Tessier N, Païta L, Pillot B, Gouriou Y, Villedieu C, Harhous Z, Paccalet A, Crola Da Silva C, Ovize M, Bidaux G, Ducreux S, Van Coppenolle F. Acute Induction of Translocon-Mediated Ca 2+ Leak Protects Cardiomyocytes Against Ischemia/Reperfusion Injury. Cells 2020; 9:cells9051319. [PMID: 32466308 PMCID: PMC7290748 DOI: 10.3390/cells9051319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
During myocardial infarction, dysregulation of Ca2+ homeostasis between the reticulum, mitochondria, and cytosol occurs in cardiomyocytes and leads to cell death. Ca2+ leak channels are thought to be key regulators of the reticular Ca2+ homeostasis and cell survival. The present study aimed to determine whether a particular reticular Ca2+ leak channel, the translocon, also known as translocation channel, could be a relevant target against ischemia/reperfusion-mediated heart injury. To achieve this objective, we first used an intramyocardial adenoviral strategy to express biosensors in order to assess Ca2+ variations in freshly isolated adult mouse cardiomyocytes to show that translocon is a functional reticular Ca2+ leak channel. Interestingly, translocon activation by puromycin mobilized a ryanodine receptor (RyR)-independent reticular Ca2+ pool and did not affect the excitation–concentration coupling. Second, puromycin pretreatment decreased mitochondrial Ca2+ content and slowed down the mitochondrial permeability transition pore (mPTP) opening and the rate of cytosolic Ca2+ increase during hypoxia. Finally, this translocon pre-activation also protected cardiomyocytes after in vitro hypoxia reoxygenation and reduced infarct size in mice submitted to in vivo ischemia-reperfusion. Altogether, our report emphasizes the role of translocon in cardioprotection and highlights a new paradigm in cardioprotection by functionally uncoupling the RyR-dependent Ca2+ stores and translocon-dependent Ca2+ stores.
Collapse
Affiliation(s)
- Ribal Al-Mawla
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Mallory Ducrozet
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Nolwenn Tessier
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Lucille Païta
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Bruno Pillot
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Yves Gouriou
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Camille Villedieu
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Zeina Harhous
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Alexandre Paccalet
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Claire Crola Da Silva
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Michel Ovize
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
- Cardiovascular functional explorations, Louis Pradel hospital, Hospices Civils de Lyon, 69677 Lyon, France
| | - Gabriel Bidaux
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| | - Sylvie Ducreux
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
- Correspondence:
| | - Fabien Van Coppenolle
- Univ Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69500 Bron, France; (R.A.-M.); (M.D.); (N.T.); (L.P.); (B.P.); (Y.G.); (C.V.); (Z.H.); (A.P.); (C.C.D.S.); (M.O.); (G.B.); (F.V.C.)
- Hospices Civils de Lyon, Groupement Hospitalier EST, Département de Cardiologie, IHU-OPERA Bâtiment B13, 69500 Bron, France
| |
Collapse
|
33
|
Vo TS, Kim YS, Ngo DN, Ngo DH. Myricetin from Rhodomyrtus tomentosa (Aiton) Hassk fruits attenuates inflammatory responses in histamine-exposed endothelial cells. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Fan L, Li L, Yu X, Liang Z, Cai T, Chen Y, Xu Y, Hu T, Wu L, Lin L. Jianpiyifei II Granules Suppress Apoptosis of Bronchial Epithelial Cells in Chronic Obstructive Pulmonary Disease via Inhibition of the Reactive Oxygen Species-Endoplasmic Reticulum Stress-Ca 2+ Signaling Pathway. Front Pharmacol 2020; 11:581. [PMID: 32425799 PMCID: PMC7204496 DOI: 10.3389/fphar.2020.00581] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Jianpiyifei II granules (JPYF II), a herbal formula, are used for the treatment of chronic obstructive pulmonary disease (COPD) in Guangdong Provincial Hospital of Chinese Medicine. The protective effects of JPYF II against bronchial epithelial cell apoptosis in mice exposed to cigarette smoke (CS) and apoptosis of human bronchial epithelial cell lines (BEAS-2B and 16-HBE) stimulated with cigarette smoke extract (CSE) were investigated. Mice were exposed to CS generated from four cigarettes/day for 30 days and administered a dose of JPYF II (0.75, 1.5, and 3 g/kg/d) from the 3rd week of CS exposure. In mice exposed to CS, JPYF II significantly inhibited CS-induced apoptosis and overexpression of endoplasmic reticulum (ER) stress-related markers in bronchial epithelial cells of the lung tissues. In CSE-stimulated BEAS-2B and 16-HBE cells, JPYF II attenuated apoptosis and cell cycle arrest in the G0/G1 phase. Mechanistically, CSE initially induced intracellular reactive oxygen species (ROS) production, which then triggered ER stress, leading to the release of Ca2+ from ER inositol trisphosphate receptor (IP3R)-mediated stores and finally cell death. Treatment with JPYF II resulted in a significant reduction in CSE-induced apoptosis through interruption of the ROS-ER stress-Ca2+ signaling pathway. Therefore, the results of this study have revealed the underlying mechanism of action of JPYF II in the treatment of COPD.
Collapse
Affiliation(s)
- Long Fan
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Leng Li
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuhua Yu
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziyao Liang
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tiantian Cai
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanbin Chen
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinji Xu
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Hu
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Wu
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Lin
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Rosa N, Sneyers F, Parys JB, Bultynck G. Type 3 IP 3 receptors: The chameleon in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:101-148. [PMID: 32247578 DOI: 10.1016/bs.ircmb.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), intracellular calcium (Ca2+) release channels, fulfill key functions in cell death and survival processes, whose dysregulation contributes to oncogenesis. This is essentially due to the presence of IP3Rs in microdomains of the endoplasmic reticulum (ER) in close proximity to the mitochondria. As such, IP3Rs enable efficient Ca2+ transfers from the ER to the mitochondria, thus regulating metabolism and cell fate. This review focuses on one of the three IP3R isoforms, the type 3 IP3R (IP3R3), which is linked to proapoptotic ER-mitochondrial Ca2+ transfers. Alterations in IP3R3 expression have been highlighted in numerous cancer types, leading to dysregulations of Ca2+ signaling and cellular functions. However, the outcome of IP3R3-mediated Ca2+ transfers for mitochondrial function is complex with opposing effects on oncogenesis. IP3R3 can either suppress cancer by promoting cell death and cellular senescence or support cancer by driving metabolism, anabolic processes, cell cycle progression, proliferation and invasion. The aim of this review is to provide an overview of IP3R3 dysregulations in cancer and describe how such dysregulations alter critical cellular processes such as proliferation or cell death and survival. Here, we pose that the IP3R3 isoform is not only linked to proapoptotic ER-mitochondrial Ca2+ transfers but might also be involved in prosurvival signaling.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium.
| |
Collapse
|
36
|
Gruszczynska-Biegala J, Strucinska K, Maciag F, Majewski L, Sladowska M, Kuznicki J. STIM Protein-NMDA2 Receptor Interaction Decreases NMDA-Dependent Calcium Levels in Cortical Neurons. Cells 2020; 9:E160. [PMID: 31936514 PMCID: PMC7017226 DOI: 10.3390/cells9010160] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/05/2020] [Accepted: 01/07/2020] [Indexed: 01/08/2023] Open
Abstract
Neuronal Store-Operated Ca2+ Entry (nSOCE) plays an essential role in refilling endoplasmic reticulum Ca2+ stores and is critical for Ca2+-dependent neuronal processes. SOCE sensors, STIM1 and STIM2, can activate Orai, TRP channels and AMPA receptors, and inhibit voltage-gated channels in the plasma membrane. However, the link between STIM, SOCE, and NMDA receptors, another key cellular entry point for Ca2+ contributing to synaptic plasticity and excitotoxicity, remains unclear. Using Ca2+ imaging, we demonstrated that thapsigargin-induced nSOCE was inhibited in rat cortical neurons following NMDAR inhibitors. Blocking nSOCE by its inhibitor SKF96365 enhanced NMDA-driven [Ca2+]i. Modulating STIM protein level through overexpression or shRNA inhibited or activated NMDA-evoked [Ca2+]i, respectively. Using proximity ligation assays, immunofluorescence, and co-immunoprecipitation methods, we discovered that thapsigargin-dependent effects required interactions between STIMs and the NMDAR2 subunits. Since STIMs modulate NMDAR-mediated Ca2+ levels, we propose targeting this mechanism as a novel therapeutic strategy against neuropathological conditions that feature NMDA-induced Ca2+ overload as a diagnostic criterion.
Collapse
Affiliation(s)
- Joanna Gruszczynska-Biegala
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
- Molecular Biology Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Klaudia Strucinska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
| | - Filip Maciag
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
| | - Lukasz Majewski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
| | - Maria Sladowska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology in Warsaw, 02-109 Warsaw, Poland; (K.S.); (F.M.); (L.M.); (M.S.); (J.K.)
| |
Collapse
|
37
|
Sarco-Endoplasmic Reticulum Calcium Release Model Based on Changes in the Luminal Calcium Content. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:337-370. [DOI: 10.1007/978-3-030-12457-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
Sterea AM, El Hiani Y. The Role of Mitochondrial Calcium Signaling in the Pathophysiology of Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:747-770. [PMID: 31646533 DOI: 10.1007/978-3-030-12457-1_30] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The pioneering work of Richard Altman on the presence of mitochondria in cells set in motion a field of research dedicated to uncovering the secrets of the mitochondria. Despite limitations in studying the structure and function of the mitochondria, advances in our understanding of this organelle prompted the development of potential treatments for various diseases, from neurodegenerative conditions to muscular dystrophy and cancer. As the powerhouses of the cell, the mitochondria represent the essence of cellular life and as such, a selective advantage for cancer cells. Much of the function of the mitochondria relies on Ca2+ homeostasis and the presence of effective Ca2+ signaling to maintain the balance between mitochondrial function and dysfunction and subsequently, cell survival. Ca2+ regulates the mitochondrial respiration rate which in turn increases ATP synthesis, but too much Ca2+ can also trigger the mitochondrial apoptosis pathway; however, cancer cells have evolved mechanisms to modulate mitochondrial Ca2+ influx and efflux in order to sustain their metabolic demand and ensure their survival. Therefore, targeting the mitochondrial Ca2+ signaling involved in the bioenergetic and apoptotic pathways could serve as potential approaches to treat cancer patients. This chapter will review the role of Ca2+ signaling in mediating the function of the mitochondria and its involvement in health and disease with special focus on the pathophysiology of cancer.
Collapse
Affiliation(s)
- Andra M Sterea
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada
| | - Yassine El Hiani
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
39
|
Lee AR, Seo MJ, Kim J, Lee DM, Kim IY, Yoon MJ, Hoon H, Choi KS. Lercanidipine Synergistically Enhances Bortezomib Cytotoxicity in Cancer Cells via Enhanced Endoplasmic Reticulum Stress and Mitochondrial Ca 2+ Overload. Int J Mol Sci 2019; 20:ijms20246112. [PMID: 31817163 PMCID: PMC6941136 DOI: 10.3390/ijms20246112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022] Open
Abstract
The proteasome inhibitor (PI), bortezomib (Btz), is effective in treating multiple myeloma and mantle cell lymphoma, but not solid tumors. In this study, we show for the first time that lercanidipine (Ler), an antihypertensive drug, enhances the cytotoxicity of various PIs, including Btz, carfilzomib, and ixazomib, in many solid tumor cell lines by inducing paraptosis, which is accompanied by severe vacuolation derived from the endoplasmic reticulum (ER) and mitochondria. We found that Ler potentiates Btz-mediated ER stress and ER dilation, possibly due to misfolded protein accumulation, in MDA-MB 435S cells. In addition, the combination of Btz and Ler triggers mitochondrial Ca2+ overload, critically contributing to mitochondrial dilation and subsequent paraptotic events, including mitochondrial membrane potential loss and ER dilation. Taken together, our results suggest that a combined regimen of PI and Ler may effectively kill cancer cells via structural and functional perturbations of the ER and mitochondria.
Collapse
Affiliation(s)
- A Reum Lee
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - Min Ji Seo
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - Jin Kim
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - Dong Min Lee
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
| | - In Young Kim
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
| | - Mi Jin Yoon
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
| | - Hur Hoon
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
- Department of Surgery, Ajou University School of Medicine, Suwon 16499, Korea
| | - Kyeong Sook Choi
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (A.R.L.); (M.J.S.); (J.K.); (D.M.L.); (I.Y.K.); (M.J.Y.)
- Department of Biomedical Science, Ajou University Graduate School of Medicine, Suwon 16499, Korea;
- Correspondence: ; Tel.: +82-31-219-4552, Fax: +82-31-219-5059
| |
Collapse
|
40
|
Schlüter A, Rossberger S, Dannehl D, Janssen JM, Vorwald S, Hanne J, Schultz C, Mauceri D, Engelhardt M. Dynamic Regulation of Synaptopodin and the Axon Initial Segment in Retinal Ganglion Cells During Postnatal Development. Front Cell Neurosci 2019; 13:318. [PMID: 31417359 PMCID: PMC6682679 DOI: 10.3389/fncel.2019.00318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
A key component allowing a neuron to function properly within its dynamic environment is the axon initial segment (AIS), the site of action potential generation. In visual cortex, AIS of pyramidal neurons undergo periods of activity-dependent structural plasticity during development. However, it remains unknown how AIS morphology is organized during development for downstream cells in the visual pathway (retinal ganglion cells; RGCs) and whether AIS retain the ability to dynamically adjust to changes in network state. Here, we investigated the maturation of AIS in RGCs during mouse retinal development, and tested putative activity-dependent mechanisms by applying visual deprivation with a focus on the AIS-specific cisternal organelle (CO), a presumed Ca2+-store. Whole-mount retinae from wildtype and Thy1-GFP transgenic mice were processed for multi-channel immunofluorescence using antibodies against AIS scaffolding proteins ankyrin-G, βIV-spectrin and the CO marker synaptopodin (synpo). Confocal microscopy in combination with morphometrical analysis of AIS length and position as well as synpo cluster size was performed. Data indicated that a subset of RGC AIS contains synpo clusters and that these show significant dynamic regulation in size during development as well as after visual deprivation. Using super resolution microscopy, we addressed the subcellular localization of synpo in RGC axons. Similar to cortical neurons, RGCs show a periodic distribution of AIS scaffolding proteins. A previously reported scaffold-deficient nanodomain correlating with synpo localization is not evident in all RGC AIS. In summary, our work demonstrates a dynamic regulation of both the AIS and synpo in RGCs during retinal development and after visual deprivation, providing first evidence that the AIS and CO in RGCs can undergo structural plasticity in response to changes in network activity.
Collapse
Affiliation(s)
- Annabelle Schlüter
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Sabrina Rossberger
- Kirchhoff-Institute for Physics, Applied Optics, Heidelberg University, Heidelberg Germany
| | - Dominik Dannehl
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jan Maximilian Janssen
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Silke Vorwald
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Christian Schultz
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Center for Biomedical Research and Medical Technology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother 2019; 118:109249. [PMID: 31351428 DOI: 10.1016/j.biopha.2019.109249] [Citation(s) in RCA: 294] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022] Open
Abstract
The endoplasmic reticulum (ER) acts as a moving organelle with many important cellular functions. As the ER lacks sufficient nutrients under pathological conditions leading to uncontrolled protein synthesis, aggregation of unfolded/misfolded proteins in the ER lumen causes the unfolded protein response (UPR) to be activated. Chronic ER stress produces endogenous or exogenous damage to cells and activates UPR, which leads to impaired intracellular calcium and redox homeostasis. The UPR is capable of recognizing the accumulation of unfolded proteins in the ER. The protein response enhances the ability of the ER to fold proteins and causes apoptosis when the function of the ER fails to return to normal. In different malignancies, ER stress can effectively induce the occurrence of autophagy in cells because malignant tumor cells need to re-use their organelles to maintain growth. Autophagy simultaneously counteracts ER stress-induced ER expansion and has the effect of enhancing cell viability and non-apoptotic death. Oxidative stress also affects mitochondrial function of important proteins through protein overload. Mitochondrial reactive oxygen species (ROS) are produced by calcium-enhanced ER release. The accumulation of toxic substances in ER and mitochondria in mitochondria destroys basic organelle function. It is known that sustained ER stress can also trigger an inflammatory response through the UPR pathway. Inflammatory response is thought to be associated with tumor development. This review discusses the emerging links between UPR responses and autophagy, oxidative stress, and inflammatory response signals in ER stress, as well as the potential development of targeting this multifaceted signaling pathway in various cancers.
Collapse
|
42
|
Calcium Signaling Pathways: Key Pathways in the Regulation of Obesity. Int J Mol Sci 2019; 20:ijms20112768. [PMID: 31195699 PMCID: PMC6600289 DOI: 10.3390/ijms20112768] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Nowadays, high epidemic obesity-triggered hypertension and diabetes seriously damage social public health. There is now a general consensus that the body's fat content exceeding a certain threshold can lead to obesity. Calcium ion is one of the most abundant ions in the human body. A large number of studies have shown that calcium signaling could play a major role in increasing energy consumption by enhancing the metabolism and the differentiation of adipocytes and reducing food intake through regulating neuronal excitability, thereby effectively decreasing the occurrence of obesity. In this paper, we review multiple calcium signaling pathways, including the IP3 (inositol 1,4,5-trisphosphate)-Ca2+ (calcium ion) pathway, the p38-MAPK (mitogen-activated protein kinase) pathway, and the calmodulin binding pathway, which are involved in biological clock, intestinal microbial activity, and nerve excitability to regulate food intake, metabolism, and differentiation of adipocytes in mammals, resulting in the improvement of obesity.
Collapse
|
43
|
Li J, Liu C, Li Y, Zheng Q, Xu Y, Liu B, Sun W, Li Y, Ji S, Liu M, Zhang J, Zhao D, Du R, Liu Z, Zhong G, Sun C, Wang Y, Song J, Zhang S, Qin J, Ling S, Wang X, Li Y. TMCO1-mediated Ca 2+ leak underlies osteoblast functions via CaMKII signaling. Nat Commun 2019; 10:1589. [PMID: 30962442 PMCID: PMC6453895 DOI: 10.1038/s41467-019-09653-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Transmembrane and coiled-coil domains 1 (TMCO1) is a recently identified Ca2+ leak channel in the endoplasmic reticulum. TMCO1 dysfunction in humans is associated with dysmorphism, mental retardation, glaucoma and the occurrence of cancer. Here we show an essential role of TMCO1 in osteogenesis mediated by local Ca2+/CaMKII signaling in osteoblasts. TMCO1 levels were significantly decreased in bone from both osteoporosis patients and bone-loss mouse models. Tmco1−/− mice exhibited loss of bone mass and altered microarchitecture characteristic of osteoporosis. In the absence of TMCO1, decreased HDAC4 phosphorylation resulted in nuclear enrichment of HADC4, which leads to deacetylation and degradation of RUNX2, the master regulator of osteogenesis. We further demonstrate that TMCO1-mediated Ca2+ leak provides local Ca2+ signals to activate the CaMKII-HDAC4-RUNX2 signaling axis. The establishment of TMCO1 as a pivotal player in osteogenesis uncovers a novel potential therapeutic target for ameliorating osteoporosis. TMCO1 is a recently described endoplasmic reticular Ca2+ channel. Here, the authors show it is important for osteoblast function and bone formation in mice, and identify a novel pathway linking local increases in Ca2+ at the ER surface with the posttranslational modification of RUNX2.
Collapse
Affiliation(s)
- Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, China
| | - Caizhi Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qiaoxia Zheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Youjia Xu
- The Second Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Beibei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Weijia Sun
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yuan Li
- The Second Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shuhui Ji
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jing Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Cuiwei Sun
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Yanqing Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Jinping Song
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, 710032, Xi'an, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, 100871, China.
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| |
Collapse
|
44
|
Lang S, Nguyen D, Pfeffer S, Förster F, Helms V, Zimmermann R. Functions and Mechanisms of the Human Ribosome-Translocon Complex. Subcell Biochem 2019; 93:83-141. [PMID: 31939150 DOI: 10.1007/978-3-030-28151-9_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The membrane of the endoplasmic reticulum (ER) in human cells harbors the protein translocon, which facilitates membrane insertion and translocation of almost every newly synthesized polypeptide targeted to organelles of the secretory pathway. The translocon comprises the polypeptide-conducting Sec61 channel and several additional proteins, which are associated with the heterotrimeric Sec61 complex. This ensemble of proteins facilitates ER targeting of precursor polypeptides, Sec61 channel opening and closing, and modification of precursor polypeptides in transit through the Sec61 complex. Recently, cryoelectron tomography of translocons in native ER membranes has given unprecedented insights into the architecture and dynamics of the native, ribosome-associated translocon and the Sec61 channel. These structural data are discussed in light of different Sec61 channel activities including ribosome receptor function, membrane insertion or translocation of newly synthesized polypeptides as well as the possible roles of the Sec61 channel as a passive ER calcium leak channel and regulator of ATP/ADP exchange between cytosol and ER.
Collapse
Affiliation(s)
- Sven Lang
- Competence Center for Molecular Medicine, Saarland University Medical School, Building 44, 66421, Homburg, Germany.
| | - Duy Nguyen
- Center for Bioinformatics, Saarland University, 66041, Saarbrücken, Germany
| | - Stefan Pfeffer
- Department of Molecular Structural Biology, Max-Planck Institute of Biochemistry, 82152, Martinsried, Germany
- ZMBH, 69120, Heidelberg, Germany
| | - Friedrich Förster
- Department of Molecular Structural Biology, Max-Planck Institute of Biochemistry, 82152, Martinsried, Germany
- Center for Biomolecular Research, Utrecht University, 3584 CH, Utrecht, The Netherlands
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, 66041, Saarbrücken, Germany
| | - Richard Zimmermann
- Competence Center for Molecular Medicine, Saarland University Medical School, Building 44, 66421, Homburg, Germany
| |
Collapse
|
45
|
Tominaga T, Sharma I, Fujita Y, Doi T, Wallner AK, Kanwar YS. Myo-inositol oxygenase accentuates renal tubular injury initiated by endoplasmic reticulum stress. Am J Physiol Renal Physiol 2018; 316:F301-F315. [PMID: 30539651 DOI: 10.1152/ajprenal.00534.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Besides oxidant stress, endoplasmic reticulum (ER) stress has been implicated in the pathogenesis of various metabolic disorders affecting the kidney. These two forms of stresses are not mutually exclusive to each other and may operate by a feedback loop in worsening the cellular injury. To attest to this contention, studies were performed to assess whether in such a setting, there is worsening of tubulointerstitial injury. We employed tunicamycin as a model of ER stress and used tubular cells and mice overexpressing myo-inositol oxygenase (MIOX), an enzyme involved in glycolytic events with excessive generation of ROS. Concomitant treatment of tunicamycin and transfection of cells with MIOX-pcDNA led to a marked generation of ROS, which was reduced by MIOX-siRNA. Likewise, an accentuated expression of ER stress sensors, GRP78, XBP1, and CHOP, was observed, which was reduced with MIOX-siRNA. These sensors were markedly elevated in MIOX-TG mice compared with WT treated with tunicamycin. This was accompanied with marked deterioration of tubular morphology, along with impairment of renal functions. Interestingly, minimal damage and elevation of ER stressors was observed in MIOX-KO mice. Downstream events that were more adversely affected in MIOX-TG mice included accentuated expression of proapoptogenic proteins, proinflammatory cytokines, and extracellular matrix constituents, although expression of these molecules was unaffected in MIOX-KO mice. Also, their tunicamycin-induced accentuated expression in tubular cells was notably reduced with MIOX-siRNA. These studies suggest that the biology of MIOX-induced oxidant stress and tunicamycin-induced ER stress are interlinked, and both of the events may feed into each other to amplify the tubulointerstitial injury.
Collapse
Affiliation(s)
- Tatsuya Tominaga
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Isha Sharma
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Yui Fujita
- Department of Nephrology, Tokushima University , Tokushima , Japan
| | - Toshio Doi
- Department of Nephrology, Tokushima University , Tokushima , Japan
| | - Aryana K Wallner
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| | - Yashpal S Kanwar
- Departments of Pathology and Medicine, Northwestern University , Chicago, Illinois
| |
Collapse
|
46
|
5-Azido-8-ethynyl-NAADP: A bifunctional, clickable photoaffinity probe for the identification of NAADP receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1180-1188. [PMID: 30521871 DOI: 10.1016/j.bbamcr.2018.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate is an evolutionarily conserved second messenger, which mobilizes Ca2+ from acidic stores. The molecular identity of the NAADP receptor has yet to be defined. In pursuit of isolating and identifying NAADP-binding proteins, we synthesized and characterized a bifunctional probe that incorporates both a photoactivatable crosslinking azido moiety at the 5-position of the nicotinic ring and a 'clickable' ethynyl moiety to the 8-adenosyl position in NAADP. Microinjection of this 5N3-8-ethynyl-NAADP into cultured U2OS cells induced robust Ca2+ responses. Higher concentrations of 5N3-8-ethynyl were required to elicit Ca2+ release or displace 32P-NAADP in radioligand binding experiments in sea urchin egg homogenates. In human cell extracts, incubation of 32P-5N3-8-ethynyl-NAADP followed by UV irradiation resulted in selective labeling of 23 kDa and 35 kDa proteins and photolabeling of these proteins was prevented when incubated in the presence of unlabeled NAADP. Compared to the monofunctional 32P-5N3-NAADP, the clickable 32P-5N3-8-ethynyl-NAADP demonstrated less labeling of the 23 kDa and 35 kDa proteins (~3-fold) but provided an opportunity for further enrichment through the 'clickable' ethynyl moiety. No proteins were specifically labeled by 32P-5N3-8-ethynyl-NAADP in sea urchin egg homogenate. These experiments demonstrate that 5N3-8-ethynyl-NAADP is biologically active and selectively labels putative NAADP-binding proteins in mammalian systems, evidencing a 'bifunctional' probe with utility for isolating NAADP-binding proteins.
Collapse
|
47
|
Kono T, Tong X, Taleb S, Bone RN, Iida H, Lee CC, Sohn P, Gilon P, Roe MW, Evans-Molina C. Impaired Store-Operated Calcium Entry and STIM1 Loss Lead to Reduced Insulin Secretion and Increased Endoplasmic Reticulum Stress in the Diabetic β-Cell. Diabetes 2018; 67:2293-2304. [PMID: 30131394 PMCID: PMC6198337 DOI: 10.2337/db17-1351] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 08/08/2018] [Indexed: 12/24/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a dynamic process that leads to refilling of endoplasmic reticulum (ER) Ca2+ stores through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor Stromal Interaction Molecule 1 (STIM1). Pathogenic reductions in β-cell ER Ca2+ have been observed in diabetes. However, a role for impaired SOCE in this phenotype has not been tested. We measured the expression of SOCE molecular components in human and rodent models of diabetes and found a specific reduction in STIM1 mRNA and protein levels in human islets from donors with type 2 diabetes (T2D), islets from hyperglycemic streptozotocin-treated mice, and INS-1 cells (rat insulinoma cells) treated with proinflammatory cytokines and palmitate. Pharmacologic SOCE inhibitors led to impaired islet Ca2+ oscillations and insulin secretion, and these effects were phenocopied by β-cell STIM1 deletion. STIM1 deletion also led to reduced ER Ca2+ storage and increased ER stress, whereas STIM1 gain of function rescued β-cell survival under proinflammatory conditions and improved insulin secretion in human islets from donors with T2D. Taken together, these data suggest that the loss of STIM1 and impaired SOCE contribute to ER Ca2+ dyshomeostasis under diabetic conditions, whereas efforts to restore SOCE-mediated Ca2+ transients may have the potential to improve β-cell health and function.
Collapse
Affiliation(s)
- Tatsuyoshi Kono
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
| | - Xin Tong
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Solaema Taleb
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Robert N Bone
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Hitoshi Iida
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Chih-Chun Lee
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Paul Sohn
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Patrick Gilon
- Pôle d'endocrinologie, diabète et nutrition, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium
| | - Michael W Roe
- Department of Medicine, SUNY Upstate Medical University, Syracuse, NY
| | - Carmella Evans-Molina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
48
|
Poletto V, Rosti V, Biggiogera M, Guerra G, Moccia F, Porta C. The role of endothelial colony forming cells in kidney cancer's pathogenesis, and in resistance to anti-VEGFR agents and mTOR inhibitors: A speculative review. Crit Rev Oncol Hematol 2018; 132:89-99. [PMID: 30447930 DOI: 10.1016/j.critrevonc.2018.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 08/07/2018] [Accepted: 09/08/2018] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinoma (RCC) is highly dependent on angiogenesis, due to the overactivation of the VHL/HIF/VEGF/VEGFRs axis; this justifies the marked sensitivity of this neoplasm to antiangiogenic agents which, however, ultimately fail to control tumor growth. RCC also frequently shows alterations in the mTOR signaling pathway, and mTOR inhibitors have shown a similar pattern of initial activity/late failure as pure antiangiogenic agents. Understanding mechanisms of resistance to these agents would be key to improve the outcome of our patients. Circulating endothelial cells are a family of mainly bone marrow-derived progenitors, which have been postulated to be responsible of the reactivation of angiogenesis in different tumors. In this review, we shall discuss the complex nature and function of these cells, the evidence pro and contra their contribution to tumor vascularization, especially as far as RCC is concerned, and their possible role in determining resistance to presently available treatments.
Collapse
Affiliation(s)
- Valentina Poletto
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy.
| | - Francesco Moccia
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, via Forlanini 6, 27100, Pavia, Italy.
| | - Camillo Porta
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy; present address: Department of Internal Medicine, University of Pavia, and Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, via S. Maugeri 10, 27100 Pavia, Italy.
| |
Collapse
|
49
|
Agrawal A, Suryakumar G, Rathor R. Role of defective Ca 2+ signaling in skeletal muscle weakness: Pharmacological implications. J Cell Commun Signal 2018; 12:645-659. [PMID: 29982883 DOI: 10.1007/s12079-018-0477-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/27/2018] [Indexed: 01/19/2023] Open
Abstract
The misbehaving attitude of Ca2+ signaling pathways could be the probable reason in many muscular disorders such as myopathies, systemic disorders like hypoxia, sepsis, cachexia, sarcopenia, heart failure, and dystrophy. The present review throws light upon the calcium flux regulating signaling channels like ryanodine receptor complex (RyR1), SERCA (Sarco-endoplasmic Reticulum Calcium ATPase), DHPR (Dihydropyridine Receptor) or Cav1.1 and Na+/Ca2+ exchange pump in detail and how remodelling of these channels contribute towards disturbed calcium homeostasis. Understanding these pathways will further provide an insight for establishing new therapeutic approaches for the prevention and treatment of muscle atrophy under stress conditions, targeting calcium ion channels and associated regulatory proteins.
Collapse
Affiliation(s)
- Akanksha Agrawal
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Geetha Suryakumar
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Richa Rathor
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
50
|
Schlüter A, Del Turco D, Deller T, Gutzmann A, Schultz C, Engelhardt M. Structural Plasticity of Synaptopodin in the Axon Initial Segment during Visual Cortex Development. Cereb Cortex 2018; 27:4662-4675. [PMID: 28922860 DOI: 10.1093/cercor/bhx208] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
The axon initial segment (AIS) is essential for action potential generation. Recently, the AIS was identified as a site of neuronal plasticity. A subpopulation of AIS in cortical principal neurons contains stacks of endoplasmic reticulum (ER) forming the cisternal organelle (CO). The function of this organelle is poorly understood, but roles in local Ca2+-trafficking and AIS plasticity are discussed. To investigate whether the presence and/or the size of COs are linked to the development and maturation of AIS of cortical neurons, we analyzed the relationship between COs and the AIS during visual cortex development under control and visual deprivation conditions. In wildtype mice, immunolabeling for synaptopodin, ankyrin-G, and ßIV-spectrin were employed to label COs and the AIS, respectively. Dark rearing resulted in an increase in synaptopodin cluster sizes, suggesting a homeostatic function of the CO in this cellular compartment. In line with this observation, synaptopodin-deficient mice lacking the CO showed AIS shortening in the dark. Collectively, these data demonstrate that the CO is an essential part of the AIS machinery required for AIS plasticity during a critical developmental period of the visual cortex.
Collapse
Affiliation(s)
- Annabelle Schlüter
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany.,Department of Neurobiology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Annika Gutzmann
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| | - Christian Schultz
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167 Mannheim, Germany
| |
Collapse
|