1
|
Huang W, Gong Y, Yan L. ER Stress, the Unfolded Protein Response and Osteoclastogenesis: A Review. Biomolecules 2023; 13:1050. [PMID: 37509086 PMCID: PMC10377020 DOI: 10.3390/biom13071050] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Endoplasmic reticulum (ER) stress and its adaptive mechanism, the unfolded protein response (UPR), are triggered by the accumulation of unfolded and misfolded proteins. During osteoclastogenesis, a large number of active proteins are synthesized. When an imbalance in the protein folding process occurs, it causes osteoclasts to trigger the UPR. This close association has led to the role of the UPR in osteoclastogenesis being increasingly explored. In recent years, several studies have reported the role of ER stress and UPR in osteoclastogenesis and bone resorption. Here, we reviewed the relevant literature and discussed the UPR signaling cascade response, osteoclastogenesis-related signaling pathways, and the role of UPR in osteoclastogenesis and bone resorption in detail. It was found that the UPR signal (PERK, CHOP, and IRE1-XBP1) promoted the expression of the receptor activator of the nuclear factor-kappa B ligand (RANKL) in osteoblasts and indirectly enhanced osteoclastogenesis. IRE1 promoted osteoclastogenesis via promoting NF-κB, MAPK signaling, or the release of pro-inflammatory factors (IL-6, IL-1β, and TNFα). CREBH promoted osteoclast differentiation by promoting NFATc1 expression. The PERK signaling pathway also promoted osteoclastogenesis through NF-κB and MAPK signaling pathways, autophagy, and RANKL secretion from osteoblasts. However, salubrinal (an inhibitor of eIF2α dephosphorylation that upregulated p-eIF2α expression) directly inhibited osteoclastogenesis by suppressing NFATc1 expression and indirectly promoted osteoclastogenesis by promoting RANKL secretion from osteoblasts. Therefore, the specific effects and mechanisms of p-PERK and its downstream signaling on osteoclastogenesis still need further experiments to confirm. In addition, the exact role of ATF6 and BiP in osteoclastogenesis also required further exploration. In conclusion, our detailed and systematic review provides some references for the next step to fully elucidate the relationship between UPR and osteoclastogenesis, intending to provide new insights for the treatment of diseases caused by osteoclast over-differentiation, such as osteoporosis.
Collapse
Affiliation(s)
- Wangli Huang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yining Gong
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Liang Yan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| |
Collapse
|
2
|
Yang F, Zhao J, Chen G, Han H, Hu S, Wang N, Wang J, Chen Y, Zhou Z, Dai B, Hou Y, Liu Y. Design, synthesis, and evaluation of hydrazones as dual inhibitors of ryanodine receptors and acetylcholinesterases for Alzheimer's disease. Bioorg Chem 2023; 133:106432. [PMID: 36841050 DOI: 10.1016/j.bioorg.2023.106432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Alzheimer's disease (AD) implicates neuronal loss, plaque and neurofibrillary tangle formation, and disturbed neuronal Ca2+ homeostasis, which leads to severe dementia, memory loss, as well as thinking and behavioral perturbations that could ultimately lead to death. Calcium dysregulation and low acetylcholine levels are two main mechanisms implicated in Alzheimer's disease progression. Simultaneous inhibition of calcium oscillations (store overload-induced Ca2+ release [SOICR]) and acetylcholinesterase (AChE) by a single molecule may bring a new breath of hope for AD treatment. Here, we described some dantrolene derivatives as dual inhibitors of the ryanodine receptor and AChE. Two series of acylhydrazone/sulfonylhydrazone derivatives with aromaticgroup were designed and synthesized. In this study, the target compounds were evaluated for their ability to inhibit SOICR and AChE in vitro, using dantrolene and donepezil as positive controls. Compound 22a exhibited excellent and balanced inhibitory potency against SOICR (inhibition (%) = 90.1, IC50 = 0.162 μM) and AChE (inhibition (%) = 93.5, IC50 = 0.372 μM). Docking simulations showed that several preferred compounds could bind to the active sites of both the proteins, further validating the rationality of the design strategy. Potential therapeutic effects in AD were evaluated using the Barnes maze and Morris water maze tests, which demonstrated that compound 22a significantly improved memory and cognitive behavior in AD model mice. Moreover, it was also found that compound 22a could enhance synaptic strength by measuring hippocampal long-term potentiation (LTP) in brain slices. These results suggested that the introduction of a sulfonyl-hydrazone scaffold and aromatic substitution to dantrolene derivatives provided a useful template for the development of potential chemical entities against AD.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Jiangang Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Guang Chen
- Department of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Hao Han
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Shuang Hu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Ningwei Wang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Junqin Wang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yuzhen Chen
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Zihao Zhou
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Baozhu Dai
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yunlei Hou
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| |
Collapse
|
3
|
Han B, Zhen F, Zheng XS, Hu J, Chen XS. Systematic analysis of the expression and prognostic value of ITPR1 and correlation with tumor infiltrating immune cells in breast cancer. BMC Cancer 2022; 22:297. [PMID: 35313846 PMCID: PMC8939201 DOI: 10.1186/s12885-022-09410-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND ITPR1 is a key gene for autophagy, but its biological function is still unclear, and there are few studies on the correlation between ITPR1 gene expression and the occurrence and development of breast cancer. METHODS Analyze the expression of ITPR1 through online databases such as Oncomine and TIMER. Kaplan-Meier plotter and other databases were used to evaluate the impact of ITPR1 on clinical prognosis. The expression of ITPR1 in analysis of 145 cases of breast cancer and 30 cases of adjacent normal tissue was detected by Immunohistochemistry. Statistical analysis was used to evaluate the clinical relevance and prognostic significance of abnormally expressed proteins. And the Western Blot was used to detect the expression of ITPR1 between breast cancer tissues and cells. The TIMER database studied the relationship between ITPR1 and cancer immune infiltration. And used the ROC plotter database to predict the response of ITPR1 to chemotherapy, endocrine therapy and anti-HER2 therapy in patients with breast cancer. RESULTS Compared with normal breast samples, ITPR1 was significantly lower in patients with breast cancer. And the increased expression of ITPR1 mRNA was closely related to longer overall survival (OS), distant metastasis free survival (DMFS), disease specific survival (DSS) and relapse free survival (RFS) in breast cancer. And the expression level of ITPR1 was higher in patients treated with chemotherapy than untreated patients. In addition, the expression of ITPR1 was positively correlated with related gene markers of immune cells in different types of breast cancer, especially with BRCA basal tissue breast cancer. CONCLUSION ITPR1 was lower expressed in breast cancer. The higher expression of ITPR1 suggested favorable prognosis for patients. ITPR1 was related to the level of immune infiltration, especially in BRCA-Basal patients. All research results indicated that ITPR1 might affect breast cancer prognosis and participate in immune regulation. In short, ITPR1 might be a potential target for breast cancer therapy.
Collapse
Affiliation(s)
- Bing Han
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Fang Zhen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Xiu-Shuang Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, 150001, China
| | - Jing Hu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Xue-Song Chen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
4
|
Zhang K, Cremers MM, Wiedemann S, Poitz DM, Pfluecke C, Heinzel FR, Pieske B, Adams V, Schauer A, Winzer R, Strasser RH, Linke A, Quick S, Heidrich FM. Spatio-temporal regulation of calpain activity after experimental myocardial infarction in vivo. Biochem Biophys Rep 2021; 28:101162. [PMID: 34761128 PMCID: PMC8566776 DOI: 10.1016/j.bbrep.2021.101162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/25/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background Calpains are calcium activated cysteine proteases that play a pivotal role in the pathophysiology of cardiac remodeling. Methods Here, we performed left anterior descending coronary artery ligation in rats as a model for ischemic systolic heart failure and examined the time- and region-specific regulation of calpain-1 and calpain-2 in the left ventricular myocardium. Results Following anterior wall myocardial infarction, calpain activity was significantly increased restricted to the ischemic anterior area at days 1, 5 and 14. No changes in calpain activity at neither time point were detected in the borderzone and remote posterior area of the left ventricle. Of note, calpain activity in the infarcted anterior myocardium was regulated differentially in the acute vs. subacute and chronic phase. In the acute phase, calpain translocation to the plasma membrane and attenuation of the expression of its endogenous inhibitor, calpastatin, were identified as the driving forces. In the subacute and chronic phase, calpain activity was regulated at the level of protein expression that was shown to be essentially independent of transcriptional activity. Conclusions We conclude that myocardial infarction leads to a distinct calpain regulation pattern in the left ventricular myocardium that is region specific and time dependent. Considering the results from our previous studies, a spatio-temporal interaction between calpains and calcium dependent natriuretic peptide production in the infarcted myocardium is possible. General significance Our results shed more light in the differential regulation of calpain activity in the myocardium and might aid in the development of targeted post-infarct and/or heart failure therapeutics.
Collapse
Key Words
- AGTR1, angiotensin II receptor type 1
- Calcium
- Calpain
- Calpain-1
- Calpain-2
- Calpastatin
- Experimental myocardial infarction
- InsP3, inositol 1,4,5-trisphosphate
- InsP3R, inositol 1,4,5-trisphopshate receptor
- LAD, left anterior descending
- LVEDD, left ventricular enddiastolic diameter
- LVEF, left ventricular ejection fraction
- LVESD, left ventricular endsystolic diameter
- NF-ĸB, nuclear factor kappa B
- NT pro-ANP, N-terminal pro atrial natriuretic peptide
- SBDP, spectrin breakdown products
Collapse
Affiliation(s)
- Kun Zhang
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Melissa M Cremers
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Stephan Wiedemann
- Helios Klinikum Pirna, Department of Internal Medicine and Cardiology, Pirna, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christian Pfluecke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Volker Adams
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Antje Schauer
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Robert Winzer
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl Gustav Carus University, Technische Universität Dresden, Dresden, Germany
| | - Ruth H Strasser
- Technische Universität Dresden, Medical Faculty, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Silvio Quick
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Felix M Heidrich
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
5
|
Woll KA, Van Petegem F. Calcium Release Channels: Structure and Function of IP3 Receptors and Ryanodine Receptors. Physiol Rev 2021; 102:209-268. [PMID: 34280054 DOI: 10.1152/physrev.00033.2020] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ca2+-release channels are giant membrane proteins that control the release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. The two members, ryanodine receptors (RyRs) and inositol-1,4,5-trisphosphate Receptors (IP3Rs), are evolutionarily related and are both activated by cytosolic Ca2+. They share a common architecture, but RyRs have evolved additional modules in the cytosolic region. Their massive size allows for the regulation by tens of proteins and small molecules, which can affect the opening and closing of the channels. In addition to Ca2+, other major triggers include IP3 for the IP3Rs, and depolarization of the plasma membrane for a particular RyR subtype. Their size has made them popular targets for study via electron microscopic methods, with current structures culminating near 3Å. The available structures have provided many new mechanistic insights int the binding of auxiliary proteins and small molecules, how these can regulate channel opening, and the mechanisms of disease-associated mutations. They also help scrutinize previously proposed binding sites, as some of these are now incompatible with the structures. Many questions remain around the structural effects of post-translational modifications, additional binding partners, and the higher-order complexes these channels can make in situ. This review summarizes our current knowledge about the structures of Ca2+-release channels and how this informs on their function.
Collapse
Affiliation(s)
- Kellie A Woll
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Complicity of α-synuclein oligomer and calcium dyshomeostasis in selective neuronal vulnerability in Lewy body disease. Arch Pharm Res 2021; 44:564-573. [PMID: 34114191 PMCID: PMC8254713 DOI: 10.1007/s12272-021-01334-6] [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: 02/22/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022]
Abstract
α-Synuclein oligomers and Ca2+ dyshomeostasis have been thoroughly investigated with respect to the pathogenesis of Lewy body disease (LBD). In LBD, α-synuclein oligomers exhibit a neuron-specific cytoplasmic distribution. Highly active neurons and neurons with a high Ca2+ burden are prone to damage in LBD. The neuronal vulnerability may be determined by transneuronal axonal transmission of the pathological processes; however, this hypothesis seems inconsistent with pathological findings that neurons anatomically connected to LBD-vulnerable neurons, such as neurons in the ventral tegmentum, are spared in LBD. This review focuses on and discusses the crucial roles played by α-synuclein oligomers and Ca2+ dyshomeostasis in early intraneural pathophysiology in LBD-vulnerable neurons. A challenging view is proposed on the synergy between retrograde transport of α-synuclein and vesicular Ca release, whereby neuronal vulnerability is propagated backward along repeatedly activated signaling pathway.
Collapse
|
7
|
Le Berre L, Chesneau M, Danger R, Dubois F, Chaussabel D, Garand M, Brouard S. Connection of BANK1, Tolerance, Regulatory B cells, and Apoptosis: Perspectives of a Reductionist Investigation. Front Immunol 2021; 12:589786. [PMID: 33815360 PMCID: PMC8015775 DOI: 10.3389/fimmu.2021.589786] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/06/2021] [Indexed: 12/07/2022] Open
Abstract
BANK1 transcript is upregulated in whole blood after kidney transplantation in tolerant patients. In comparison to patients with rejection, tolerant patients display higher level of regulatory B cells (Bregs) expressing granzyme B (GZMB+) that have the capability to prevent effector T cells proliferation. However, BANK1 was found to be decreased in these GZMB+ Bregs. In this article, we investigated seven different transcriptomic studies and mined the literature in order to make link between BANK1, tolerance and Bregs. As for GZMB+ Bregs, we found that BANK1 was decreased in other subtypes of Bregs, including IL10+ and CD24hiCD38hi transitional regulatory B cells, along with BANK1 was down-regulated in activated/differentiated B cells, as in CD40-activated B cells, in leukemia and plasma cells. Following a reductionist approach, biological concepts were extracted from BANK1 literature and allowed us to infer association between BANK1 and immune signaling pathways, as STAT1, FcγRIIB, TNFAIP3, TRAF6, and TLR7. Based on B cell signaling literature and expression data, we proposed a role of BANK1 in B cells of tolerant patients that involved BCR, IP3R, and PLCG2, and a link with the apoptosis pathways. We confronted these data with our experiments on apoptosis in total B cells and Bregs, and this suggests different involvement for BANK1 in these two cells. Finally, we put in perspective our own data with other published data to hypothesize two different roles for BANK1 in B cells and in Bregs.
Collapse
Affiliation(s)
- Ludmilla Le Berre
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Mélanie Chesneau
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Richard Danger
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Florian Dubois
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | | | - Mathieu Garand
- Systems Biology and Immunology, Sidra Medicine, Doha, Qatar
| | - Sophie Brouard
- CHU Nantes, Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
8
|
Ivanova H, Vervliet T, Monaco G, Terry LE, Rosa N, Baker MR, Parys JB, Serysheva II, Yule DI, Bultynck G. Bcl-2-Protein Family as Modulators of IP 3 Receptors and Other Organellar Ca 2+ Channels. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035089. [PMID: 31501195 DOI: 10.1101/cshperspect.a035089] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pro- and antiapoptotic proteins belonging to the B-cell lymphoma-2 (Bcl-2) family exert a critical control over cell-death processes by enabling or counteracting mitochondrial outer membrane permeabilization. Beyond this mitochondrial function, several Bcl-2 family members have emerged as critical modulators of intracellular Ca2+ homeostasis and dynamics, showing proapoptotic and antiapoptotic functions. Bcl-2 family proteins specifically target several intracellular Ca2+-transport systems, including organellar Ca2+ channels: inositol 1,4,5-trisphosphate receptors (IP3Rs) and ryanodine receptors (RyRs), Ca2+-release channels mediating Ca2+ flux from the endoplasmic reticulum, as well as voltage-dependent anion channels (VDACs), which mediate Ca2+ flux across the mitochondrial outer membrane into the mitochondria. Although the formation of protein complexes between Bcl-2 proteins and these channels has been extensively studied, a major advance during recent years has been elucidating the complex interaction of Bcl-2 proteins with IP3Rs. Distinct interaction sites for different Bcl-2 family members were identified in the primary structure of IP3Rs. The unique molecular profiles of these Bcl-2 proteins may account for their distinct functional outcomes when bound to IP3Rs. Furthermore, Bcl-2 inhibitors used in cancer therapy may affect IP3R function as part of their proapoptotic effect and/or as an adverse effect in healthy cells.
Collapse
Affiliation(s)
- Hristina Ivanova
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Giovanni Monaco
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Lara E Terry
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Nicolas Rosa
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Mariah R Baker
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Structural Biology Imaging Center, Houston, Texas 77030
| | - Jan B Parys
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, McGovern Medical School at The University of Texas Health Science Center at Houston, Structural Biology Imaging Center, Houston, Texas 77030
| | - David I Yule
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
9
|
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]
|
10
|
Lopez JJ, Jardin I, Albarrán L, Sanchez-Collado J, Cantonero C, Salido GM, Smani T, Rosado JA. Molecular Basis and Regulation of Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:445-469. [PMID: 31646520 DOI: 10.1007/978-3-030-12457-1_17] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous mechanism for Ca2+ influx in mammalian cells with important physiological implications. Since the discovery of SOCE more than three decades ago, the mechanism that communicates the information about the amount of Ca2+ accumulated in the intracellular Ca2+ stores to the plasma membrane channels and the nature of these channels have been matters of intense investigation and debate. The stromal interaction molecule-1 (STIM1) has been identified as the Ca2+ sensor of the intracellular Ca2+ compartments that activates the store-operated channels. STIM1 regulates two types of store-dependent channels: the Ca2+ release-activated Ca2+ (CRAC) channels, formed by Orai1 subunits, that conduct the highly Ca2+ selective current I CRAC and the cation permeable store-operated Ca2+ (SOC) channels, which consist of Orai1 and TRPC1 proteins and conduct the non-selective current I SOC. While the crystal structure of Drosophila CRAC channel has already been solved, the architecture of the SOC channels still remains unclear. The dynamic interaction of STIM1 with the store-operated channels is modulated by a number of proteins that either support the formation of the functional STIM1-channel complex or protect the cell against Ca2+ overload.
Collapse
Affiliation(s)
- Jose J Lopez
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Isaac Jardin
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain.
| | - Letizia Albarrán
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Jose Sanchez-Collado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Carlos Cantonero
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Gines M Salido
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics and Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Sevilla, Sevilla, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| |
Collapse
|
11
|
Lemos FDO, Florentino RM, Lima Filho ACM, Santos MLD, Leite MF. Inositol 1,4,5-trisphosphate receptor in the liver: Expression and function. World J Gastroenterol 2019; 25:6483-6494. [PMID: 31802829 PMCID: PMC6886013 DOI: 10.3748/wjg.v25.i44.6483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a complex organ that performs several functions to maintain homeostasis. These functions are modulated by calcium, a second messenger that regulates several intracellular events. In hepatocytes and cholangiocytes, which are the epithelial cell types in the liver, inositol 1,4,5-trisphosphate (InsP3) receptors (ITPR) are the only intracellular calcium release channels. Three isoforms of the ITPR have been described, named type 1, type 2 and type 3. These ITPR isoforms are differentially expressed in liver cells where they regulate distinct physiological functions. Changes in the expression level of these receptors correlate with several liver diseases and hepatic dysfunctions. In this review, we highlight how the expression level, modulation, and localization of ITPR isoforms in hepatocytes and cholangiocytes play a role in hepatic homeostasis and liver pathology.
Collapse
Affiliation(s)
- Fernanda de Oliveira Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Antônio Carlos Melo Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Marcone Loiola dos Santos
- Department of Cell Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - M Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
12
|
Zhang K, Wiedemann S, Dschietzig M, Cremers MM, Augstein A, Poitz DM, Quick S, Pfluecke C, Heinzel FR, Pieske B, Adams V, Linke A, Strasser RH, Heidrich FM. The infarction zone rather than the noninfarcted remodeling zone overexpresses angiotensin II receptor type 1 and is the main source of ventricular atrial natriuretic peptide. Cardiovasc Pathol 2019; 44:107160. [PMID: 31759320 DOI: 10.1016/j.carpath.2019.107160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/05/2019] [Accepted: 10/07/2019] [Indexed: 12/20/2022] Open
Abstract
Chromogranin B and inositol 1,4,5-trisphosphate-associated calcium signaling leading to increased natriuretic peptide production has been described in cardiac hypertrophy. Here, we performed left anterior descending coronary artery ligation in rats as a model for systolic heart failure and examined protein and gene expression clusters in the infarcted and noninfarcted myocardium and moreover under treatment with metoprolol. We found that atrial natriuretic peptide gene transcription was significantly more elevated in the infarcted compared with the noninfarcted myocardium. Chromogranin B, which facilitates calcium release from internal stores through the inositol 1,4,5-trisphosphate receptor, was upregulated in both areas. Interestingly, angiotensin II receptor type 1 gene transcription was significantly upregulated in the infarcted and unchanged in the noninfarcted myocardium. Nuclear factor ĸappa B as a calcium-dependent transcription factor showed increased activity in the infarction zone. The β-adrenergic axis does not seem to be involved, as metoprolol treatment did not have a significant impact on any of these results. We conclude that region-specific upregulation of angiotensin II receptor type 1 is a major factor for increased atrial natriuretic peptide production in the infarcted anterior wall. This effect is most likely achieved through inositol 1,4,5-trisphosphate-mediated cytosolic calcium increase and subsequent nuclear factor ĸappa B activation, which is a known transcription factor for natriuretic peptides.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Stephan Wiedemann
- Helios Klinikum Pirna, Department of Internal Medicine and Cardiology, Pirna, Germany
| | - Martin Dschietzig
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Melissa M Cremers
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Antje Augstein
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Silvio Quick
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Christian Pfluecke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Volker Adams
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Axel Linke
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany
| | - Ruth H Strasser
- Technische Universität Dresden, Medical Faculty, Dresden, Germany
| | - Felix M Heidrich
- Department of Internal Medicine and Cardiology, Herzzentrum Dresden at Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
13
|
Yamamoto K, Izumi Y, Arifuku M, Kume T, Sawada H. α-Synuclein oligomers mediate the aberrant form of spike-induced calcium release from IP 3 receptor. Sci Rep 2019; 9:15977. [PMID: 31685859 PMCID: PMC6828767 DOI: 10.1038/s41598-019-52135-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/12/2019] [Indexed: 01/10/2023] Open
Abstract
Emerging evidence implicates α-synuclein oligomers as potential culprits in the pathogenesis of Lewy body disease (LBD). Soluble oligomeric α-synuclein accumulation in cytoplasm is believed to modify neuronal activities and intraneural Ca2+ dynamics, which augment the metabolic burden in central neurons vulnerable to LBD, although this hypothesis remains to be fully tested. We evaluated how intracellular α-synuclein oligomers affect the neuronal excitabilities and Ca2+ dynamics of pyramidal neurons in neocortical slices from mice. Intracellular application of α-synuclein containing stable higher-order oligomers (αSNo) significantly reduced spike frequency during current injection, elongated the duration of spike afterhyperpolarization (AHP), and enlarged AHP current charge in comparison with that of α-synuclein without higher-order oligomers. This αSNo-mediated alteration was triggered by spike-induced Ca2+ release from inositol trisphosphate receptors (IP3R) functionally coupled with L-type Ca2+ channels and SK-type K+ channels. Further electrophysiological and immunochemical observations revealed that α-synuclein oligomers greater than 100 kDa were directly associated with calcium-binding protein 1, which is responsible for regulating IP3R gating. They also block Ca2+-dependent inactivation of IP3R, and trigger Ca2+-induced Ca2+ release from IP3R during multiple spikes. This aberrant machinery may result in intraneural Ca2+ dyshomeostasis and may be the molecular basis for the vulnerability of neurons in LBD brains.
Collapse
Affiliation(s)
- Kenji Yamamoto
- Department of Neurology and Clinical Research Center, National Hospital Organization Utano National Hospital, Kyoto, Japan.
| | - Yasuhiko Izumi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Kobe, Japan.,Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Monami Arifuku
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.,Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hideyuki Sawada
- Department of Neurology and Clinical Research Center, National Hospital Organization Utano National Hospital, Kyoto, Japan
| |
Collapse
|
14
|
Rathour RK, Narayanan R. Degeneracy in hippocampal physiology and plasticity. Hippocampus 2019; 29:980-1022. [PMID: 31301166 PMCID: PMC6771840 DOI: 10.1002/hipo.23139] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/27/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
Degeneracy, defined as the ability of structurally disparate elements to perform analogous function, has largely been assessed from the perspective of maintaining robustness of physiology or plasticity. How does the framework of degeneracy assimilate into an encoding system where the ability to change is an essential ingredient for storing new incoming information? Could degeneracy maintain the balance between the apparently contradictory goals of the need to change for encoding and the need to resist change towards maintaining homeostasis? In this review, we explore these fundamental questions with the mammalian hippocampus as an example encoding system. We systematically catalog lines of evidence, spanning multiple scales of analysis that point to the expression of degeneracy in hippocampal physiology and plasticity. We assess the potential of degeneracy as a framework to achieve the conjoint goals of encoding and homeostasis without cross-interferences. We postulate that biological complexity, involving interactions among the numerous parameters spanning different scales of analysis, could establish disparate routes towards accomplishing these conjoint goals. These disparate routes then provide several degrees of freedom to the encoding-homeostasis system in accomplishing its tasks in an input- and state-dependent manner. Finally, the expression of degeneracy spanning multiple scales offers an ideal reconciliation to several outstanding controversies, through the recognition that the seemingly contradictory disparate observations are merely alternate routes that the system might recruit towards accomplishment of its goals.
Collapse
Affiliation(s)
- Rahul K. Rathour
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| | - Rishikesh Narayanan
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| |
Collapse
|
15
|
Conrard L, Tyteca D. Regulation of Membrane Calcium Transport Proteins by the Surrounding Lipid Environment. Biomolecules 2019; 9:E513. [PMID: 31547139 PMCID: PMC6843150 DOI: 10.3390/biom9100513] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
Calcium ions (Ca2+) are major messengers in cell signaling, impacting nearly every aspect of cellular life. Those signals are generated within a wide spatial and temporal range through a large variety of Ca2+ channels, pumps, and exchangers. More and more evidences suggest that Ca2+ exchanges are regulated by their surrounding lipid environment. In this review, we point out the technical challenges that are currently being overcome and those that still need to be defeated to analyze the Ca2+ transport protein-lipid interactions. We then provide evidences for the modulation of Ca2+ transport proteins by lipids, including cholesterol, acidic phospholipids, sphingolipids, and their metabolites. We also integrate documented mechanisms involved in the regulation of Ca2+ transport proteins by the lipid environment. Those include: (i) Direct interaction inside the protein with non-annular lipids; (ii) close interaction with the first shell of annular lipids; (iii) regulation of membrane biophysical properties (e.g., membrane lipid packing, thickness, and curvature) directly around the protein through annular lipids; and (iv) gathering and downstream signaling of several proteins inside lipid domains. We finally discuss recent reports supporting the related alteration of Ca2+ and lipids in different pathophysiological events and the possibility to target lipids in Ca2+-related diseases.
Collapse
Affiliation(s)
- Louise Conrard
- CELL Unit, de Duve Institute and Université catholique de Louvain, UCL B1.75.05, avenue Hippocrate, 75, B-1200 Brussels, Belgium
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute and Université catholique de Louvain, UCL B1.75.05, avenue Hippocrate, 75, B-1200 Brussels, Belgium.
| |
Collapse
|
16
|
Causton B, Pardo-Saganta A, Gillis J, Discipio K, Kooistra T, Rajagopal J, Xavier RJ, Cho JL, Medoff BD. CARMA3 Mediates Allergic Lung Inflammation in Response to Alternaria alternata. Am J Respir Cell Mol Biol 2019; 59:684-694. [PMID: 29958012 DOI: 10.1165/rcmb.2017-0181oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The airway epithelial cell (AEC) response to allergens helps initiate and propagate allergic inflammation in asthma. CARMA3 is a scaffold protein that mediates G protein-coupled receptor-induced NF-κB activation in airway epithelium. In this study, we demonstrate that mice with CARMA3-deficient AECs have reduced airway inflammation, as well as reduced type 2 cytokine levels in response to Alternaria alternata. These mice also have reduced production of IL-33 and IL-25, and reduced numbers of innate lymphoid cells in the lung. We also show that CARMA3-deficient human AECs have decreased production of proasthmatic mediators in response to A. alternata. Finally, we show that CARMA3 interacts with inositol 1,4,5-trisphosphate receptors in AECs, and that inhibition of CARMA3 signaling reduces A. alternata-induced intracellular calcium release. In conclusion, we show that CARMA3 signaling in AECs helps mediate A. alternata-induced allergic airway inflammation, and that CARMA3 is an important signaling molecule for type 2 immune responses in the lung.
Collapse
Affiliation(s)
- Benjamin Causton
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Ana Pardo-Saganta
- 1 Division of Pulmonary and Critical Care Medicine.,4 Center for Computational and Integrative Biology, and
| | - Jacob Gillis
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Katherine Discipio
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Tristan Kooistra
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Jayaraj Rajagopal
- 1 Division of Pulmonary and Critical Care Medicine.,4 Center for Computational and Integrative Biology, and
| | - Ramnik J Xavier
- 5 Gastrointestinal Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; and.,2 Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Josalyn L Cho
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| | - Benjamin D Medoff
- 1 Division of Pulmonary and Critical Care Medicine.,3 Center for Regenerative Medicine
| |
Collapse
|
17
|
Potapenko E, Negrão NW, Huang G, Docampo R. The acidocalcisome inositol-1,4,5-trisphosphate receptor of Trypanosoma brucei is stimulated by luminal polyphosphate hydrolysis products. J Biol Chem 2019; 294:10628-10637. [PMID: 31138655 DOI: 10.1074/jbc.ra119.007906] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/27/2019] [Indexed: 12/21/2022] Open
Abstract
Acidocalcisomes are acidic calcium stores rich in polyphosphate (polyP) and are present in trypanosomes and also in a diverse range of other organisms. Ca2+ is released from these organelles through a channel, inositol 1,4,5-trisphosphate receptor (TbIP3R), which is essential for growth and infectivity of the parasite Trypanosoma brucei However, the mechanism by which TbIP3R controls Ca2+ release is unclear. In this work, we expressed TbIP3R in a chicken B lymphocyte cell line in which the genes for all three vertebrate IP3Rs were stably ablated (DT40-3KO). We show that IP3-mediated Ca2+ release depends on Ca2+ but not on ATP concentration and is inhibited by heparin, caffeine, and 2-aminomethoxydiphenyl borate (2-APB). Excised patch clamp recordings from nuclear membranes of DT40 cells expressing only TbIP3R disclosed that luminal inorganic orthophosphate (Pi) or pyrophosphate (PPi), and neutral or alkaline pH can stimulate IP3-generated currents. In contrast, polyP or acidic pH did not induce these currents, and nuclear membranes obtained from cells expressing rat IP3R were unresponsive to polyP or its hydrolysis products. Our results are consistent with the notion that polyP hydrolysis products within acidocalcisomes or alkalinization of their luminal pH activate TbIP3R and Ca2+ release. We conclude that TbIP3R is well-adapted to its role as the major Ca2+ release channel of acidocalcisomes in T. brucei.
Collapse
Affiliation(s)
| | - Núria W Negrão
- From the Center for Tropical and Emerging Global Diseases and.,Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| | - Guozhong Huang
- From the Center for Tropical and Emerging Global Diseases and
| | - Roberto Docampo
- From the Center for Tropical and Emerging Global Diseases and .,Department of Cellular Biology, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
18
|
Ashhad S, Narayanan R. Stores, Channels, Glue, and Trees: Active Glial and Active Dendritic Physiology. Mol Neurobiol 2019; 56:2278-2299. [PMID: 30014322 PMCID: PMC6394607 DOI: 10.1007/s12035-018-1223-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
Glial cells and neuronal dendrites were historically assumed to be passive structures that play only supportive physiological roles, with no active contribution to information processing in the central nervous system. Research spanning the past few decades has clearly established this assumption to be far from physiological realities. Whereas the discovery of active channel conductances and their localized plasticity was the turning point for dendritic structures, the demonstration that glial cells release transmitter molecules and communicate across the neuroglia syncytium through calcium wave propagation constituted path-breaking discoveries for glial cell physiology. An additional commonality between these two structures is the ability of calcium stores within their endoplasmic reticulum (ER) to support active propagation of calcium waves, which play crucial roles in the spatiotemporal integration of information within and across cells. Although there have been several demonstrations of regulatory roles of glial cells and dendritic structures in achieving common physiological goals such as information propagation and adaptability through plasticity, studies assessing physiological interactions between these two active structures have been few and far. This lacuna is especially striking given the strong connectivity that is known to exist between these two structures through several complex and tightly intercoupled mechanisms that also recruit their respective ER structures. In this review, we present brief overviews of the parallel literatures on active dendrites and active glial physiology and make a strong case for future studies to directly assess the strong interactions between these two structures in regulating physiology and pathophysiology of the brain.
Collapse
Affiliation(s)
- Sufyan Ashhad
- Department of Neurobiology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
19
|
Apasu JE, Schuette D, LaRanger R, Steinle JA, Nguyen LD, Grosshans HK, Zhang M, Cai WL, Yan Q, Robert ME, Mak M, Ehrlich BE. Neuronal calcium sensor 1 (NCS1) promotes motility and metastatic spread of breast cancer cells in vitro and in vivo. FASEB J 2018; 33:4802-4813. [PMID: 30592625 DOI: 10.1096/fj.201802004r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Increased levels of the calcium-binding protein neuronal calcium sensor 1 (NCS1) predict an unfavorable patient outcome in several aggressive cancers, including breast and liver tumors. Previous studies suggest that NCS1 overexpression facilitates metastatic spread of these cancers. To investigate this hypothesis, we explored the effects of NCS1 overexpression on cell proliferation, survival, and migration patterns in vitro in 2- and 3-dimensional (2/3-D). Furthermore, we translated our results into an in vivo mouse xenograft model. Cell-based proliferation assays were used to demonstrate the effects of overexpression of NCS1 on growth rates. In vitro colony formation and wound healing experiments were performed and 3-D migration dynamics were studied using collagen gels. Nude mice were injected with breast cancer cells to monitor NCS1-dependent metastasis formation over time. We observed that increased NCS1 levels do not change cellular growth rates, but do significantly increase 2- and 3-D migration dynamics in vitro. Likewise, NCS1-overexpressing cells have an increased capacity to form distant metastases and demonstrate better survival and less necrosis in vivo. We found that NCS1 preferentially localizes to the leading edge of cells and overexpression increases the motility of cancer cells. Furthermore, this phenotype is correlated with an increased number of metastases in a xenograft model. These results lay the foundation for exploring the relevance of an NCS1-mediated pathway as a metastatic biomarker and as a target for pharmacologic interventions.-Apasu, J. E., Schuette, D., LaRanger, R., Steinle, J. A., Nguyen, L. D., Grosshans, H. K., Zhang, M., Cai, W. L., Yan, Q., Robert, M. E., Mak, M., Ehrlich, B. E. Neuronal calcium sensor 1 (NCS1) promotes motility and metastatic spread of breast cancer cells in vitro and in vivo.
Collapse
Affiliation(s)
- Jonathan E Apasu
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Daniel Schuette
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Ryan LaRanger
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA; and
| | - Julia A Steinle
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Lien D Nguyen
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | | | - Meiling Zhang
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Wesley L Cai
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Qin Yan
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA; and
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
20
|
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
|
21
|
Cryo-EM reveals ligand induced allostery underlying InsP 3R channel gating. Cell Res 2018; 28:1158-1170. [PMID: 30470765 PMCID: PMC6274648 DOI: 10.1038/s41422-018-0108-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/02/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023] Open
Abstract
Inositol-1,4,5-trisphosphate receptors (InsP3Rs) are cation channels that mobilize Ca2+ from intracellular stores in response to a wide range of cellular stimuli. The paradigm of InsP3R activation is the coupled interplay between binding of InsP3 and Ca2+ that switches the ion conduction pathway between closed and open states to enable the passage of Ca2+ through the channel. However, the molecular mechanism of how the receptor senses and decodes ligand-binding signals into gating motion remains unknown. Here, we present the electron cryo-microscopy structure of InsP3R1 from rat cerebellum determined to 4.1 Å resolution in the presence of activating concentrations of Ca2+ and adenophostin A (AdA), a structural mimetic of InsP3 and the most potent known agonist of the channel. Comparison with the 3.9 Å-resolution structure of InsP3R1 in the Apo-state, also reported herein, reveals the binding arrangement of AdA in the tetrameric channel assembly and striking ligand-induced conformational rearrangements within cytoplasmic domains coupled to the dilation of a hydrophobic constriction at the gate. Together, our results provide critical insights into the mechanistic principles by which ligand-binding allosterically gates InsP3R channel.
Collapse
|
22
|
Sampieri A, Santoyo K, Asanov A, Vaca L. Association of the IP3R to STIM1 provides a reduced intraluminal calcium microenvironment, resulting in enhanced store-operated calcium entry. Sci Rep 2018; 8:13252. [PMID: 30185837 PMCID: PMC6125598 DOI: 10.1038/s41598-018-31621-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/15/2018] [Indexed: 11/21/2022] Open
Abstract
The involvement of inositol trisphosphate receptor (IP3R) in modulating store-operated calcium entry (SOCE) was established many years ago. Nevertheless, the molecular mechanism responsible for this observation has not been elucidated to this date. In the present study we show that IP3R associates to STIM1 upon depletion of the endoplasmic reticulum (ER) by activation of the inositol trisphosphate signaling cascade via G-protein coupled receptors. IP3R-STIM1 association results in enhanced STIM1 puncta formation and larger Orai-mediated whole-cell currents as well as increased calcium influx. Depleting the ER with a calcium ATPase inhibitor (thapsigargin, TG) does not induce IP3R-STIM1 association, indicating that this association requires an active IP3R. The IP3R-STIM1 association is only observed after IP3R activation, as evidenced by FRET experiments and co-immunoprecipitation assays. ER intraluminal calcium measurements using Mag-Fluo-4 showed enhanced calcium depletion when IP3R is overexpressed. A STIM1-GCaMP fusion protein indicates that STIM1 detects lower calcium concentrations near its EF-hand domain when IP3R is overexpressed when compared with the fluorescence reported by a GCaMP homogenously distributed in the ER lumen (ER-GCaMP). All these data together strongly suggest that activation of inositol trisphosphate signaling cascade induces the formation of the IP3R-STIM1 complex. The activated IP3R provides a reduced intraluminal calcium microenvironment near STIM1, resulting in enhanced activation of Orai currents and SOCE.
Collapse
Affiliation(s)
- Alicia Sampieri
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiología Celular. Universidad Nacional Autonoma de México, Ciudad de México, Mexico
| | - Karla Santoyo
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiología Celular. Universidad Nacional Autonoma de México, Ciudad de México, Mexico
| | | | - Luis Vaca
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiología Celular. Universidad Nacional Autonoma de México, Ciudad de México, Mexico.
| |
Collapse
|
23
|
Schuette D, Moore LM, Robert ME, Taddei TH, Ehrlich BE. Hepatocellular Carcinoma Outcome Is Predicted by Expression of Neuronal Calcium Sensor 1. Cancer Epidemiol Biomarkers Prev 2018; 27:1091-1100. [PMID: 29789326 PMCID: PMC8465775 DOI: 10.1158/1055-9965.epi-18-0167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/06/2018] [Accepted: 05/17/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. There is an urgent demand for prognostic biomarkers that facilitate early tumor detection, as the incidence of HCC has tripled in the United States in the last three decades. Biomarkers to identify populations at risk would have significant impact on survival. We recently found that expression of Neuronal Calcium Sensor 1 (NCS1), a Ca2+-dependent signaling molecule, predicted disease outcome in breast cancer, but its predictive value in other cancer types is unknown. This protein is potentially useful because increased NCS1 regulates Ca2+ signaling and increased Ca2+ signaling is a hallmark of metastatic cancers, conferring cellular motility and an increasingly aggressive phenotype to tumors.Methods: We explored the relationship between NCS1 expression levels and patient survival in two publicly available liver cancer cohorts and a tumor microarray using data mining strategies.Results: High NCS1 expression levels are significantly associated with worse disease outcome in Asian patients within these cohorts. In addition, a variety of Ca2+-dependent and tumor growth-promoting genes are transcriptionally coregulated with NCS1 and many of them are involved in cytoskeleton organization, suggesting that NCS1 induced dysregulated Ca2+ signaling facilitates cellular motility and metastasis.Conclusions: We found NCS1 to be a novel biomarker in HCC. Furthermore, our study identified a pharmacologically targetable signaling complex that can influence tumor progression in HCC.Impact: These results lay the foundation for using NCS1 as a prognostic biomarker in prospective cohorts of HCC patients and for further functional assessment of the characterized signaling axis. Cancer Epidemiol Biomarkers Prev; 27(9); 1091-100. ©2018 AACR.
Collapse
Affiliation(s)
- Daniel Schuette
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Lauren M Moore
- Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, Connecticut
| | - Tamar H Taddei
- Department of Medicine (Digestive Diseases), Yale University, New Haven, Connecticut
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, New Haven, Connecticut.
| |
Collapse
|
24
|
Li G, Che H, Wu WY, Jie LJ, Xiao GS, Wang Y, Li GR. Bradykinin-mediated Ca 2+ signalling regulates cell growth and mobility in human cardiac c-Kit + progenitor cells. J Cell Mol Med 2018; 22:4688-4699. [PMID: 30117680 PMCID: PMC6156395 DOI: 10.1111/jcmm.13706] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/27/2018] [Indexed: 01/05/2023] Open
Abstract
Our recent study showed that bradykinin increases cell cycling progression and migration of human cardiac c‐Kit+ progenitor cells by activating pAkt and pERK1/2 signals. This study investigated whether bradykinin‐mediated Ca2+ signalling participates in regulating cellular functions in cultured human cardiac c‐Kit+ progenitor cells using laser scanning confocal microscopy and biochemical approaches. It was found that bradykinin increased cytosolic free Ca2+ (Cai2+) by triggering a transient Ca2+ release from ER IP3Rs followed by sustained Ca2+ influx through store‐operated Ca2+ entry (SOCE) channel. Blockade of B2 receptor with HOE140 or IP3Rs with araguspongin B or silencing IP3R3 with siRNA abolished both Ca2+ release and Ca2+ influx. It is interesting to note that the bradykinin‐induced cell cycle progression and migration were not observed in cells with siRNA‐silenced IP3R3 or the SOCE component TRPC1, Orai1 or STIM1. Also the bradykinin‐induced increase in pAkt and pERK1/2 as well as cyclin D1 was reduced in these cells. These results demonstrate for the first time that bradykinin‐mediated increase in free Cai2+ via ER‐IP3R3 Ca2+ release followed by Ca2+ influx through SOCE channel plays a crucial role in regulating cell growth and migration via activating pAkt, pERK1/2 and cyclin D1 in human cardiac c‐Kit+ progenitor cells.
Collapse
Affiliation(s)
- Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| | - Hui Che
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Ling-Jun Jie
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong Pokfulam, Hong Kong, China
| |
Collapse
|
25
|
Tong BCK, Wu AJ, Li M, Cheung KH. Calcium signaling in Alzheimer's disease & therapies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1745-1760. [PMID: 30059692 DOI: 10.1016/j.bbamcr.2018.07.018] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and is characterized by the accumulation of amyloid (Aβ) plaques and neurofibrillary tangles in the brain. Much attention has been given to develop AD treatments based on the amyloid cascade hypothesis; however, none of these drugs had good efficacy at improving cognitive functions in AD patients suggesting that Aβ might not be the disease origin. Thus, there are urgent needs for the development of new therapies that target on the proximal cause of AD. Cellular calcium (Ca2+) signals regulate important facets of neuronal physiology. An increasing body of evidence suggests that age-related dysregulation of neuronal Ca2+ homeostasis may play a proximal role in the pathogenesis of AD as disrupted Ca2+ could induce synaptic deficits and promote the accumulation of Aβ plaques and neurofibrillary tangles. Given that Ca2+ disruption is ubiquitously involved in all AD pathologies, it is likely that using chemical agents or small molecules specific to Ca2+ channels or handling proteins on the plasma membrane and membranes of intracellular organelles to correct neuronal Ca2+ dysregulation could open up a new approach to AD prevention and treatment. This review summarizes current knowledge on the molecular mechanisms linking Ca2+ dysregulation with AD pathologies and discusses the possibility of correcting neuronal Ca2+ disruption as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Benjamin Chun-Kit Tong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Aston Jiaxi Wu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - King-Ho Cheung
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| |
Collapse
|
26
|
Distelhorst CW. Targeting Bcl-2-IP 3 receptor interaction to treat cancer: A novel approach inspired by nearly a century treating cancer with adrenal corticosteroid hormones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1795-1804. [PMID: 30053503 DOI: 10.1016/j.bbamcr.2018.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
Bcl-2 inhibits cell death by at least two different mechanisms. On the one hand, its BH3 domain binds to pro-apoptotic proteins such as Bim and prevents apoptosis induction. On the other hand, the BH4 domain of Bcl-2 binds to the inositol 1,4,5-trisphosphate receptor (IP3R), preventing Ca2+ signals that mediate cell death. In normal T-cells, Bcl-2 levels increase during the immune response, protecting against cell death, and then decline as apoptosis ensues and the immune response dissipates. But in many cancers Bcl-2 is aberrantly expressed and exploited to prevent cell death by inhibiting IP3R-mediated Ca2+ elevation. This review summarizes what is known about the mechanism of Bcl-2's control over IP3R-mediated Ca2+ release and cell death induction. Early insights into the role of Ca2+ elevation in corticosteroid-mediated cell death serves as a model for how targeting IP3R-mediated Ca2+ elevation can be a highly effective therapeutic approach for different types of cancer. Moreover, the successful development of ABT-199 (Venetoclax), a small molecule targeting the BH3 domain of Bcl-2 but without effects on Ca2+, serves as proof of principle that targeting Bcl-2 can be an effective therapeutic approach. BIRD-2, a synthetic peptide that inhibits Bcl-2-IP3R interaction, induces cell death induction in ABT-199 (Venetoclax)-resistant cancer models, attesting to the value of developing therapeutic agents that selectively target Bcl-2-IP3R interaction, inducing Ca2+-mediated cell death.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Case Western University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States of America.
| |
Collapse
|
27
|
Kerkhofs M, Giorgi C, Marchi S, Seitaj B, Parys JB, Pinton P, Bultynck G, Bittremieux M. Alterations in Ca 2+ Signalling via ER-Mitochondria Contact Site Remodelling in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:225-254. [PMID: 28815534 DOI: 10.1007/978-981-10-4567-7_17] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inter-organellar contact sites establish microdomains for localised Ca2+-signalling events. One of these microdomains is established between the ER and the mitochondria. Importantly, the so-called mitochondria-associated ER membranes (MAMs) contain, besides structural proteins and proteins involved in lipid exchange, several Ca2+-transport systems, mediating efficient Ca2+ transfer from the ER to the mitochondria. These Ca2+ signals critically control several mitochondrial functions, thereby impacting cell metabolism, cell death and survival, proliferation and migration. Hence, the MAMs have emerged as critical signalling hubs in physiology, while their dysregulation is an important factor that drives or at least contributes to oncogenesis and tumour progression. In this book chapter, we will provide an overview of the role of the MAMs in cell function and how alterations in the MAM composition contribute to oncogenic features and behaviours.
Collapse
Affiliation(s)
- Martijn Kerkhofs
- Laboratory Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), KU Leuven, Campus Gasthuisberg O&N 1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Bruno Seitaj
- Laboratory Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), KU Leuven, Campus Gasthuisberg O&N 1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Jan B Parys
- Laboratory Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), KU Leuven, Campus Gasthuisberg O&N 1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, University of Ferrara, Ferrara, Italy
| | - Geert Bultynck
- Laboratory Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), KU Leuven, Campus Gasthuisberg O&N 1 Box 802, Herestraat 49, 3000, Leuven, Belgium.
| | - Mart Bittremieux
- Laboratory Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), KU Leuven, Campus Gasthuisberg O&N 1 Box 802, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
28
|
Fouqué A, Lepvrier E, Debure L, Gouriou Y, Malleter M, Delcroix V, Ovize M, Ducret T, Li C, Hammadi M, Vacher P, Legembre P. The apoptotic members CD95, BclxL, and Bcl-2 cooperate to promote cell migration by inducing Ca(2+) flux from the endoplasmic reticulum to mitochondria. Cell Death Differ 2016; 23:1702-16. [PMID: 27367565 PMCID: PMC5041197 DOI: 10.1038/cdd.2016.61] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/26/2016] [Accepted: 06/03/2016] [Indexed: 12/16/2022] Open
Abstract
Metalloprotease-processed CD95L (cl-CD95L) is a soluble cytokine that implements a PI3K/Ca(2+) signaling pathway in triple-negative breast cancer (TNBC) cells. Accordingly, high levels of cl-CD95L in TNBC women correlate with poor prognosis, and administration of this ligand in an orthotopic xenograft mouse model accelerates the metastatic dissemination of TNBC cells. The molecular mechanism underlying CD95-mediated cell migration remains unknown. Here, we present genetic and pharmacologic evidence that the anti-apoptotic molecules BclxL and Bcl-2 and the pro-apoptotic factors BAD and BID cooperate to promote migration of TNBC cells stimulated with cl-CD95L. BclxL was distributed in both endoplasmic reticulum (ER) and mitochondrion membranes. The mitochondrion-localized isoform promoted cell migration by interacting with voltage-dependent anion channel 1 to orchestrate Ca(2+) transfer from the ER to mitochondria in a BH3-dependent manner. Mitochondrial Ca(2+) uniporter contributed to this flux, which favored ATP production and cell migration. In conclusion, this study reveals a novel molecular mechanism controlled by BclxL to promote cancer cell migration and supports the use of BH3 mimetics as therapeutic options not only to kill tumor cells but also to prevent metastatic dissemination in TNBCs.
Collapse
Affiliation(s)
- A Fouqué
- Inserm ER440-Oncogenesis, Stress and Signaling, Equipe Labellisée Ligue Contre Le Cancer, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Centre Eugène Marquis, Inserm ERL440-OSS, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Université de Rennes-1, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
| | - E Lepvrier
- Inserm ER440-Oncogenesis, Stress and Signaling, Equipe Labellisée Ligue Contre Le Cancer, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Centre Eugène Marquis, Inserm ERL440-OSS, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Université de Rennes-1, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
- Laboratoire Commun, OncoTrial UMS Biosit/Biotrial 7-9 Rue Jean-Louis Bertrand, Rennes 35000, France
| | - L Debure
- Inserm ER440-Oncogenesis, Stress and Signaling, Equipe Labellisée Ligue Contre Le Cancer, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Centre Eugène Marquis, Inserm ERL440-OSS, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Université de Rennes-1, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
| | - Y Gouriou
- Inserm U1060, CarMeN, Université Claude Bernard Lyon 1, 8 Avenue Rockfeller, Lyon 69373, France
| | - M Malleter
- Inserm ER440-Oncogenesis, Stress and Signaling, Equipe Labellisée Ligue Contre Le Cancer, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Centre Eugène Marquis, Inserm ERL440-OSS, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Université de Rennes-1, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
- Laboratoire Commun, OncoTrial UMS Biosit/Biotrial 7-9 Rue Jean-Louis Bertrand, Rennes 35000, France
| | - V Delcroix
- Université de Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
- Inserm U1218, Institut Bergonié, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - M Ovize
- Inserm U1060, CarMeN, Université Claude Bernard Lyon 1, 8 Avenue Rockfeller, Lyon 69373, France
- Hospices Civils de Lyon, Hôpital Louis Pradel, Services D'explorations Fonctionnelles Cardiovasculaires et CIC de Lyon, Lyon 69394, France
| | - T Ducret
- Université de Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
- Inserm U1045, Centre de Recherche Cardiothoracique de Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
| | - C Li
- Molecular Targets Group, James Graham Brown Cancer Center, Departments of Medicine, and Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - M Hammadi
- Université de Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
- Inserm U1218, Institut Bergonié, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - P Vacher
- Université de Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
- Inserm U1218, Institut Bergonié, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - P Legembre
- Inserm ER440-Oncogenesis, Stress and Signaling, Equipe Labellisée Ligue Contre Le Cancer, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Centre Eugène Marquis, Inserm ERL440-OSS, Rue Bataille Flandres Dunkerque, Rennes 35042, France
- Université de Rennes-1, 2 Avenue du Prof. Léon Bernard, Rennes 35043, France
| |
Collapse
|
29
|
Abstract
The role of cytosolic Ca(2+) on the kinetics of Inositol 1,4,5-triphosphate receptors (IP3Rs) and on the dynamics of IP3R-mediated Ca(2+) signals has been studied at large both experimentally and by modeling. The role of luminal Ca(2+) has not been investigated with that much detail although it has been found that it is relevant for signal termination in the case of Ca(2+) release through ryanodine receptors. In this work we present the results of observing the dynamics of luminal and cytosolic Ca(2+) simultaneously in Xenopus laevis oocytes. Combining observations and modeling we conclude that there is a rapid mechanism that guarantees the availability of free Ca(2+) in the lumen even when a relatively large Ca(2+) release is evoked. Comparing the dynamics of cytosolic and luminal Ca(2+) during a release, we estimate that they are consistent with a 80% of luminal Ca(2+) being buffered. The rapid availability of free luminal Ca(2+) correlates with the observation that the lumen occupies a considerable volume in several regions across the images.
Collapse
|
30
|
Shaikh S, Troncoso R, Criollo A, Bravo-Sagua R, García L, Morselli E, Cifuentes M, Quest AFG, Hill JA, Lavandero S. Regulation of cardiomyocyte autophagy by calcium. Am J Physiol Endocrinol Metab 2016; 310:E587-E596. [PMID: 26884385 PMCID: PMC4835942 DOI: 10.1152/ajpendo.00374.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/10/2016] [Indexed: 11/22/2022]
Abstract
Calcium signaling plays a crucial role in a multitude of events within the cardiomyocyte, including cell cycle control, growth, apoptosis, and autophagy. With respect to calcium-dependent regulation of autophagy, ion channels and exchangers, receptors, and intracellular mediators play fundamental roles. In this review, we discuss calcium-dependent regulation of cardiomyocyte autophagy, a lysosomal mechanism that is often cytoprotective, serving to defend against disease-related stress and nutrient insufficiency. We also highlight the importance of the subcellular distribution of calcium and related proteins, interorganelle communication, and other key signaling events that govern cardiomyocyte autophagy.
Collapse
Affiliation(s)
- Soni Shaikh
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lorena García
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
| | - Eugenia Morselli
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Cifuentes
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
| | - Joseph A Hill
- Departments of Internal Medicine (Cardiology Division) and
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sergio Lavandero
- Advanced Center for Chronic Disease and Center for Molecular Studies of the Cell, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile;
- Departamento de Bioquimica y Biologia Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; and
- Departments of Internal Medicine (Cardiology Division) and
| |
Collapse
|
31
|
Alzayady KJ, Wang L, Chandrasekhar R, Wagner LE, Van Petegem F, Yule DI. Defining the stoichiometry of inositol 1,4,5-trisphosphate binding required to initiate Ca2+ release. Sci Signal 2016; 9:ra35. [PMID: 27048566 DOI: 10.1126/scisignal.aad6281] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) are tetrameric intracellular Ca(2+)-release channels with each subunit containing a binding site for IP3in the amino terminus. We provide evidence that four IP3molecules are required to activate the channel under diverse conditions. Comparing the concentration-response relationship for binding and Ca(2+)release suggested that IP3Rs are maximally occupied by IP3before substantial Ca(2+)release occurs. We showed that ligand binding-deficient subunits acted in a dominant-negative manner when coexpressed with wild-type monomers in the chicken immune cell line DT40-3KO, which lacks all three genes encoding IP3R subunits, and confirmed the same effect in an IP3R-null human cell line (HEK-3KO) generated by CRISPR/Cas9 technology. Using dimeric and tetrameric concatenated IP3Rs with increasing numbers of binding-deficient subunits, we addressed the obligate ligand stoichiometry. The concatenated IP3Rs with four ligand-binding sites exhibited Ca(2+)release and electrophysiological properties of native IP3Rs. However, IP3failed to activate IP3Rs assembled from concatenated dimers consisting of one binding-competent and one binding-deficient mutant subunit. Similarly, IP3Rs containing two monomers of IP3R2short, an IP3binding-deficient splice variant, were nonfunctional. Concatenated tetramers containing only three binding-competent ligand-binding sites were nonfunctional under a wide range of activating conditions. These data provide definitive evidence that IP3-induced Ca(2+)release only occurs when each IP3R monomer within the tetramer is occupied by IP3, thereby ensuring fidelity of Ca(2+)release.
Collapse
Affiliation(s)
- Kamil J Alzayady
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Liwei Wang
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Rahul Chandrasekhar
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Larry E Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
32
|
Prole DL, Taylor CW. Inositol 1,4,5-trisphosphate receptors and their protein partners as signalling hubs. J Physiol 2016; 594:2849-66. [PMID: 26830355 PMCID: PMC4887697 DOI: 10.1113/jp271139] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/06/2015] [Indexed: 01/26/2023] Open
Abstract
Inositol 1,4,5‐trisphosphate receptors (IP3Rs) are expressed in nearly all animal cells, where they mediate the release of Ca2+ from intracellular stores. The complex spatial and temporal organization of the ensuing intracellular Ca2+ signals allows selective regulation of diverse physiological responses. Interactions of IP3Rs with other proteins contribute to the specificity and speed of Ca2+ signalling pathways, and to their capacity to integrate information from other signalling pathways. In this review, we provide a comprehensive survey of the proteins proposed to interact with IP3Rs and the functional effects that these interactions produce. Interacting proteins can determine the activity of IP3Rs, facilitate their regulation by multiple signalling pathways and direct the Ca2+ that they release to specific targets. We suggest that IP3Rs function as signalling hubs through which diverse inputs are processed and then emerge as cytosolic Ca2+ signals.
![]()
Collapse
Affiliation(s)
- David L Prole
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| |
Collapse
|
33
|
Tmem178 acts in a novel negative feedback loop targeting NFATc1 to regulate bone mass. Proc Natl Acad Sci U S A 2015; 112:15654-9. [PMID: 26644563 DOI: 10.1073/pnas.1511285112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Phospholipase C gamma-2 (PLCγ2)-dependent calcium (Ca(2+)) oscillations are indispensable for nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) activation and downstream gene transcription driving osteoclastogenesis during skeletal remodeling and pathological bone loss. Here we describe, to our knowledge, the first known function of transmembrane protein 178 (Tmem178), a PLCγ2 downstream target gene, as a critical modulator of the NFATc1 axis. In surprising contrast to the osteopetrotic phenotype of PLCγ2(-/-) mice, Tmem178(-/-) mice are osteopenic in basal conditions and are more susceptible to inflammatory bone loss, owing to enhanced osteoclast formation. Mechanistically, Tmem178 localizes to the ER membrane and regulates RANKL-induced Ca(2+) fluxes, thus controlling NFATc1 induction. Importantly, down-regulation of Tmem178 is observed in human CD14(+) monocytes exposed to plasma from systemic juvenile idiopathic arthritis patients. Similar to the mouse model, reduced Tmem178 expression in human cells correlates with excessive osteoclastogenesis. In sum, these findings identify an essential role for Tmem178 to maintain skeletal mass and limit pathological bone loss.
Collapse
|
34
|
Abstract
A number of chronic metabolic pathologies, including obesity, diabetes, cardiovascular disease, asthma, and cancer, cluster together to present the greatest threat to human health. As research in this field has advanced, it has become clear that unresolved metabolic inflammation, organelle dysfunction, and other cellular and metabolic stresses underlie the development of these chronic metabolic diseases. However, the relationship between these systems and pathological mechanisms is poorly understood. Here we discuss the role of cellular Ca(2+) homeostasis as a critical mechanism integrating the myriad of cellular and subcellular dysfunctional networks found in metabolic tissues such as liver and adipose tissue in the context of metabolic disease, particularly in obesity and diabetes.
Collapse
|
35
|
Oliveira AG, Guimarães ES, Andrade LM, Menezes GB, Fatima Leite M. Decoding calcium signaling across the nucleus. Physiology (Bethesda) 2015; 29:361-8. [PMID: 25180265 DOI: 10.1152/physiol.00056.2013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Calcium (Ca(2+)) is an important multifaceted second messenger that regulates a wide range of cellular events. A Ca(2+)-signaling toolkit has been shown to exist in the nucleus and to be capable of generating and modulating nucleoplasmic Ca(2+) transients. Within the nucleus, Ca(2+) controls cellular events that are different from those modulated by cytosolic Ca(2+). This review focuses on nuclear Ca(2+) signals and their role in regulating physiological and pathological processes.
Collapse
Affiliation(s)
- André G Oliveira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Erika S Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil; Molecular Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil; and
| | - Lídia M Andrade
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gustavo B Menezes
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - M Fatima Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil;
| |
Collapse
|
36
|
Kato H, Nishitoh H. Stress responses from the endoplasmic reticulum in cancer. Front Oncol 2015; 5:93. [PMID: 25941664 PMCID: PMC4403295 DOI: 10.3389/fonc.2015.00093] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/31/2015] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic organelle that is essential for multiple cellular functions. During cellular stress conditions, including nutrient deprivation and dysregulation of protein synthesis, unfolded/misfolded proteins accumulate in the ER lumen, resulting in activation of the unfolded protein response (UPR). The UPR also contributes to the regulation of various intracellular signaling pathways such as calcium signaling and lipid signaling. More recently, the mitochondria-associated ER membrane (MAM), which is a site of close contact between the ER and mitochondria, has been shown to function as a platform for various intracellular stress responses including apoptotic signaling, inflammatory signaling, the autophagic response, and the UPR. Interestingly, in cancer, these signaling pathways from the ER are often dysregulated, contributing to cancer cell metabolism. Thus, the signaling pathway from the ER may be a novel therapeutic target for various cancers. In this review, we discuss recent research on the roles of stress responses from the ER, including the MAM.
Collapse
Affiliation(s)
- Hironori Kato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| |
Collapse
|
37
|
Piegari E, Sigaut L, Ponce Dawson S. Ca²⁺ images obtained in different experimental conditions shed light on the spatial distribution of IP₃ receptors that underlie Ca²⁺ puffs. Cell Calcium 2015; 57:109-19. [PMID: 25636173 DOI: 10.1016/j.ceca.2015.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/29/2014] [Accepted: 01/01/2015] [Indexed: 10/24/2022]
Abstract
Many intracellular Ca(2+) signals involve Ca(2+) release from the endoplasmic reticulum through inositol 1,4,5-trisphosphate receptors (IP3Rs). The open probability of IP3Rs depends on cytosolic Ca(2+) so that these signals involve Ca(2+)-induced Ca(2+)-release (CICR). IP3Rs are organized in clusters. The signals they mediate are observed using single-wavelength dyes and, often, a slow Ca(2+) buffer (EGTA) is added to disrupt CICR between clusters and keep the signals spatially restricted. It is assumed that the presence of the dye or of EGTA does not alter the intra-cluster Ca(2+) dynamics. In this paper we analyze this issue combining experiments and numerical simulations. We compare the properties of local signals known as puffs observed with different dyes and EGTA concentrations. We determine that although the dye or EGTA does not alter the intra-cluster dynamics, the set of observable events is different depending on the degree of inter-cluster uncoupling of the experiment. An analysis of the observations shows that the events that are missed for insufficient inter-cluster uncoupling are those of fastest amplitude growth rate. This agrees with a spatial organization in which the largest amplitude events correspond to clusters with densely packed active IP3Rs.
Collapse
Affiliation(s)
- Estefanía Piegari
- Departamento de Física and IFIBA, CONICET, FCEyN-UBA, Ciudad Universitaria, Pabellón I, 1428 Buenos Aires, Argentina.
| | - Lorena Sigaut
- Departamento de Física and IFIBA, CONICET, FCEyN-UBA, Ciudad Universitaria, Pabellón I, 1428 Buenos Aires, Argentina
| | - Silvina Ponce Dawson
- Departamento de Física and IFIBA, CONICET, FCEyN-UBA, Ciudad Universitaria, Pabellón I, 1428 Buenos Aires, Argentina
| |
Collapse
|
38
|
Ashhad S, Johnston D, Narayanan R. Activation of InsP₃ receptors is sufficient for inducing graded intrinsic plasticity in rat hippocampal pyramidal neurons. J Neurophysiol 2014; 113:2002-13. [PMID: 25552640 DOI: 10.1152/jn.00833.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/29/2014] [Indexed: 11/22/2022] Open
Abstract
The synaptic plasticity literature has focused on establishing necessity and sufficiency as two essential and distinct features in causally relating a signaling molecule to plasticity induction, an approach that has been surprisingly lacking in the intrinsic plasticity literature. In this study, we complemented the recently established necessity of inositol trisphosphate (InsP3) receptors (InsP3R) in a form of intrinsic plasticity by asking if InsP3R activation was sufficient to induce intrinsic plasticity in hippocampal neurons. Specifically, incorporation of d-myo-InsP3 in the recording pipette reduced input resistance, maximal impedance amplitude, and temporal summation but increased resonance frequency, resonance strength, sag ratio, and impedance phase lead. Strikingly, the magnitude of plasticity in all these measurements was dependent on InsP3 concentration, emphasizing the graded dependence of such plasticity on InsP3R activation. Mechanistically, we found that this InsP3-induced plasticity depended on hyperpolarization-activated cyclic nucleotide-gated channels. Moreover, this calcium-dependent form of plasticity was critically reliant on the release of calcium through InsP3Rs, the influx of calcium through N-methyl-d-aspartate receptors and voltage-gated calcium channels, and on the protein kinase A pathway. Our results delineate a causal role for InsP3Rs in graded adaptation of neuronal response dynamics, revealing novel regulatory roles for the endoplasmic reticulum in neural coding and homeostasis.
Collapse
Affiliation(s)
- Sufyan Ashhad
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India; and
| | - Daniel Johnston
- Center for Learning and Memory, The University of Texas at Austin, Austin, Texas
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India; and
| |
Collapse
|
39
|
Ananthanarayanan M, Banales JM, Guerra MT, Spirli C, Munoz-Garrido P, Mitchell-Richards K, Tafur D, Saez E, Nathanson MH. Post-translational regulation of the type III inositol 1,4,5-trisphosphate receptor by miRNA-506. J Biol Chem 2014; 290:184-96. [PMID: 25378392 DOI: 10.1074/jbc.m114.587030] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The type III isoform of the inositol 1,4,5-trisphosphate receptor (InsP3R3) is apically localized and triggers Ca(2+) waves and secretion in a number of polarized epithelia. However, nothing is known about epigenetic regulation of this InsP3R isoform. We investigated miRNA regulation of InsP3R3 in primary bile duct epithelia (cholangiocytes) and in the H69 cholangiocyte cell line, because the role of InsP3R3 in cholangiocyte Ca(2+) signaling and secretion is well established and because loss of InsP3R3 from cholangiocytes is responsible for the impairment in bile secretion that occurs in a number of liver diseases. Analysis of the 3'-UTR of human InsP3R3 mRNA revealed two highly conserved binding sites for miR-506. Transfection of miR-506 mimics into cell lines expressing InsP3R3-3'UTR-luciferase led to decreased reporter activity, whereas co-transfection with miR-506 inhibitors led to enhanced activity. Reporter activity was abrogated in isolated mutant proximal or distal miR-506 constructs in miR-506-transfected HEK293 cells. InsP3R3 protein levels were decreased by miR-506 mimics and increased by inhibitors, and InsP3R3 expression was markedly decreased in H69 cells stably transfected with miR-506 relative to control cells. miR-506-H69 cells exhibited a fibrotic signature. In situ hybridization revealed elevated miR-506 expression in vivo in human-diseased cholangiocytes. Histamine-induced, InsP3-mediated Ca(2+) signals were decreased by 50% in stable miR-506 cells compared with controls. Finally, InsP3R3-mediated fluid secretion was significantly decreased in isolated bile duct units transfected with miR-506, relative to control IBDU. Together, these data identify miR-506 as a regulator of InsP3R3 expression and InsP3R3-mediated Ca(2+) signaling and secretion.
Collapse
Affiliation(s)
| | - Jesus M Banales
- the Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of Basque Country (UPV/EHU), CIBERehd, IKERBASQUE, AECC, 20014 San Sebastian, Spain, and the Division of Gene Therapy and Hepatology, CIMA of the University of Navarra, Ciberehd, 31009 Pamplona, Spain
| | | | | | - Patricia Munoz-Garrido
- the Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of Basque Country (UPV/EHU), CIBERehd, IKERBASQUE, AECC, 20014 San Sebastian, Spain, and
| | - Kisha Mitchell-Richards
- Pathology, Section of Digestive Diseases and the Liver Center, Yale University School of Medicine, New Haven, Connecticut 06520
| | | | - Elena Saez
- the Division of Gene Therapy and Hepatology, CIMA of the University of Navarra, Ciberehd, 31009 Pamplona, Spain
| | | |
Collapse
|
40
|
Wang DY, Abbasi C, El-Rass S, Li JY, Dawood F, Naito K, Sharma P, Bousette N, Singh S, Backx PH, Cox B, Wen XY, Liu PP, Gramolini AO. Endoplasmic reticulum resident protein 44 (ERp44) deficiency in mice and zebrafish leads to cardiac developmental and functional defects. J Am Heart Assoc 2014; 3:e001018. [PMID: 25332179 PMCID: PMC4323785 DOI: 10.1161/jaha.114.001018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endoplasmic reticulum (ER) resident protein 44 (ERp44) is a member of the protein disulfide isomerase family, is induced during ER stress, and may be involved in regulating Ca(2+) homeostasis. However, the role of ERp44 in cardiac development and function is unknown. The aim of this study was to investigate the role of ERp44 in cardiac development and function in mice, zebrafish, and embryonic stem cell (ESC)-derived cardiomyocytes to determine the underlying role of ERp44. METHODS AND RESULTS We generated and characterized ERp44(-/-) mice, ERp44 morphant zebrafish embryos, and ERp44(-/-) ESC-derived cardiomyocytes. Deletion of ERp44 in mouse and zebrafish caused significant embryonic lethality, abnormal heart development, altered Ca(2+) dynamics, reactive oxygen species generation, activated ER stress gene profiles, and apoptotic cell death. We also determined the cardiac phenotype in pressure overloaded, aortic-banded ERp44(+/-) mice: enhanced ER stress activation and increased mortality, as well as diastolic cardiac dysfunction with a significantly lower fractional shortening. Confocal and LacZ histochemical staining showed a significant transmural gradient for ERp44 in the adult heart, in which high expression of ERp44 was observed in the outer subepicardial region of the myocardium. CONCLUSIONS ERp44 plays a critical role in embryonic heart development and is crucial in regulating cardiac cell Ca(2+) signaling, ER stress, ROS-induced oxidative stress, and activation of the intrinsic mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Ding-Yan Wang
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Cynthia Abbasi
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Suzan El-Rass
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.) Keenan Research Center for Biomedical Science and Zebrafish Center for Advanced Drug Discovery, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada (S.E.R., X.Y.W.)
| | - Jamie Yuanjun Li
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Fayez Dawood
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Kotaro Naito
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Parveen Sharma
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Nicolas Bousette
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Shalini Singh
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Peter H Backx
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Brian Cox
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Xiao-Yan Wen
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.) Keenan Research Center for Biomedical Science and Zebrafish Center for Advanced Drug Discovery, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada (S.E.R., X.Y.W.)
| | - Peter P Liu
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Anthony O Gramolini
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| |
Collapse
|
41
|
Bonneau B, Popgeorgiev N, Prudent J, Gillet G. Cytoskeleton dynamics in early zebrafish development: A matter of phosphorylation? BIOARCHITECTURE 2014; 1:216-220. [PMID: 22754611 PMCID: PMC3384572 DOI: 10.4161/bioa.18116] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Early morphogenic movements are an important feature of embryonic development in vertebrates. During zebrafish gastrulation, epiboly progression is driven by the coordinated remodeling of the YSL microtubule network and F-actin cables. We recently described the implication of Nrz, an anti-apoptotic Bcl-2 homolog, in the control of the YSL cytoskeleton dynamics. Nrz knock-down induces premature actin-myosin ring formation leading to margin constriction, epiboly arrest and embryo lethality. At the molecular level, the Nrz protein controls the actin-myosin dynamics through IP3R-dependent calcium levels variation. Here, we discuss these novel findings and propose a model in which reversible phosphorylation of the Nrz/IP3R complex modulates the permeability of the IP3R calcium channel and thus may explain the Nrz-dependent control of IP3R opening required for proper epiboly completion.
Collapse
Affiliation(s)
- Benjamin Bonneau
- Université Lyon I; Centre de Recherche en Cancérologie de Lyon; Centre Léon Bérard; Lyon, France
| | | | | | | |
Collapse
|
42
|
Fos C, Becart S, Canonigo Balancio AJ, Boehning D, Altman A. Association of the EF-hand and PH domains of the guanine nucleotide exchange factor SLAT with IP₃ receptor 1 promotes Ca²⁺ signaling in T cells. Sci Signal 2014; 7:ra93. [PMID: 25270259 DOI: 10.1126/scisignal.2005565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The guanine nucleotide exchange factor SLAT (SWAP-70-like adaptor of T cells) regulates T cell activation and differentiation by enabling Ca(2+) release from intracellular stores in response to stimulation of the T cell receptor (TCR). We found a TCR-induced association between SLAT and inositol 1,4,5-trisphosphate (IP3) receptor type 1 (IP3R1). The N-terminal region of SLAT, which contains two EF-hand motifs that we determined bound Ca(2+), and the SLAT pleckstrin homology (PH) domain independently bound to IP3R1 by associating with a conserved motif within the IP3R1 ligand-binding domain. Disruption of the SLAT-IP3R1 interaction with cell-permeable, IP3R1-based fusion peptides inhibited TCR-stimulated Ca(2+) signaling, activation of the transcription factor NFAT (nuclear factor of activated T cells), and production of cytokines, suggesting that this interaction is required for optimal T cell activation. The finding that SLAT is an IP3R1-interacting protein required for T cell activation suggests that this interaction could be a potential target for a selective immunosuppressive drug.
Collapse
Affiliation(s)
- Camille Fos
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Stephane Becart
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Ann J Canonigo Balancio
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Darren Boehning
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
43
|
de Carvalho M, Eisen A, Krieger C, Swash M. Motoneuron firing in amyotrophic lateral sclerosis (ALS). Front Hum Neurosci 2014; 8:719. [PMID: 25294995 PMCID: PMC4170108 DOI: 10.3389/fnhum.2014.00719] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/27/2014] [Indexed: 01/09/2023] Open
Abstract
Amyotrophic lateral sclerosis is an inexorably progressive neurodegenerative disorder involving the classical motor system and the frontal effector brain, causing muscular weakness and atrophy, with variable upper motor neuron signs and often an associated fronto-temporal dementia. The physiological disturbance consequent on the motor system degeneration is beginning to be well understood. In this review we describe aspects of the motor cortical, neuronal, and lower motor neuron dysfunction. We show how studies of the changes in the pattern of motor unit firing help delineate the underlying pathophysiological disturbance as the disease progresses. Such studies are beginning to illuminate the underlying disordered pathophysiological processes in the disease, and are important in designing new approaches to therapy and especially for clinical trials.
Collapse
Affiliation(s)
- Mamede de Carvalho
- Institute of Physiology and Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Department of Neurosciences, Hospital Santa Maria, Faculty of Medicine, University of Lisbon Lisbon, Portugal
| | - Andrew Eisen
- Emeritus Professor of Neurology, University of British Columbia Vancouver, BC, Canada
| | - Charles Krieger
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby BC, Canada ; Department of Medicine (Neurology), University of British Columbia, Vancouver BC, Canada
| | - Michael Swash
- Institute of Physiology and Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Department of Neurosciences, Hospital Santa Maria, Faculty of Medicine, University of Lisbon Lisbon, Portugal ; Institute of Neuroscience, Barts and The London School of Medicine, Queen Mary University of London London, UK
| |
Collapse
|
44
|
Serysheva II. Toward a high-resolution structure of IP₃R channel. Cell Calcium 2014; 56:125-32. [PMID: 25159857 DOI: 10.1016/j.ceca.2014.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/01/2014] [Accepted: 08/02/2014] [Indexed: 12/11/2022]
Abstract
The ability of cells to maintain low levels of Ca(2+) under resting conditions and to create rapid and transient increases in Ca(2+) upon stimulation is a fundamental property of cellular Ca(2+) signaling mechanism. An increase of cytosolic Ca(2+) level in response to diverse stimuli is largely accounted for by the inositol 1,4,5-trisphosphate receptor (IP3R) present in the endoplasmic reticulum membranes of virtually all eukaryotic cells. Extensive information is currently available on the function of IP3Rs and their interaction with modulators. Very little, however, is known about their molecular architecture and therefore most critical issues surrounding gating of IP3R channels are still ambiguous, including the central question of how opening of the IP3R pore is initiated by IP3 and Ca(2+). Membrane proteins such as IP3R channels have proven to be exceptionally difficult targets for structural analysis due to their large size, their location in the membrane environment, and their dynamic nature. To date, a 3D structure of complete IP3R channel is determined by single-particle cryo-EM at intermediate resolution, and the best crystal structures of IP3R are limited to a soluble portion of the cytoplasmic region representing ∼15% of the entire channel protein. Together these efforts provide the important structural information for this class of ion channels and serve as the basis for further studies aiming at understanding of the IP3R function.
Collapse
Affiliation(s)
- Irina I Serysheva
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
45
|
Signore S, Sorrentino A, Ferreira-Martins J, Kannappan R, Shafaie M, Del Ben F, Isobe K, Arranto C, Wybieralska E, Webster A, Sanada F, Ogórek B, Zheng H, Liu X, del Monte F, D'Alessandro DA, Wunimenghe O, Michler RE, Hosoda T, Goichberg P, Leri A, Kajstura J, Anversa P, Rota M. Response to letter regarding article "Inositol 1,4,5-trisphosphate receptors and human left ventricular myocytes". Circulation 2014; 129:e510-1. [PMID: 24868002 DOI: 10.1161/circulationaha.114.009347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sergio Signore
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrea Sorrentino
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - João Ferreira-Martins
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ramaswamy Kannappan
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mehrdad Shafaie
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Fabio Del Ben
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kazuya Isobe
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Christian Arranto
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ewa Wybieralska
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrew Webster
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Fumihiro Sanada
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Barbara Ogórek
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hanqiao Zheng
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Xiaoxia Liu
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Federica del Monte
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - David A D'Alessandro
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Oriyanhan Wunimenghe
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Robert E Michler
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Toru Hosoda
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Polina Goichberg
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Annarosa Leri
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jan Kajstura
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Piero Anversa
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marcello Rota
- Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
46
|
Feng X, Krogh KA, Wu CY, Lin YW, Tsai HC, Thayer SA, Wei LN. Receptor-interacting protein 140 attenuates endoplasmic reticulum stress in neurons and protects against cell death. Nat Commun 2014; 5:4487. [PMID: 25066731 PMCID: PMC4200015 DOI: 10.1038/ncomms5487] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/23/2014] [Indexed: 12/29/2022] Open
Abstract
Inositol 1, 4, 5-trisphosphate receptor (IP3R)-mediated Ca(2+) release from the endoplasmic reticulum (ER) triggers many physiological responses in neurons, and when uncontrolled can cause ER stress that contributes to neurological disease. Here we show that the unfolded protein response (UPR) in neurons induces rapid translocation of nuclear receptor-interacting protein 140 (RIP140) to the cytoplasm. In the cytoplasm, RIP140 localizes to the ER by binding to the IP3R. The carboxyl-terminal RD4 domain of RIP140 interacts with the carboxyl-terminal gate-keeping domain of the IP3R. This molecular interaction disrupts the IP3R's 'head-tail' interaction, thereby suppressing channel opening and attenuating IP3R-mediated Ca(2+) release. This contributes to a rapid suppression of the ER stress response and provides protection from apoptosis in both hippocampal neurons in vitro and in an animal model of ER stress. Thus, RIP140 translocation to the cytoplasm is an early response to ER stress and provides protection against neuronal death.
Collapse
Affiliation(s)
- Xudong Feng
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kelly A. Krogh
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Cheng-Ying Wu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yi-Wei Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Hong-Chieh Tsai
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Department of Neurosurgery, Chang-Gung Memorial Hospital and University, Tao-Yuan, Taiwan, R.O.C
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Heidrich FM, Strasser RH, Wiedemann S. Letter by Heidrich et al regarding article, "Inositol 1,4,5-trisphosphate receptors and human left ventricular myocytes". Circulation 2014; 129:e509. [PMID: 24868001 DOI: 10.1161/circulationaha.113.007565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Felix M Heidrich
- Technische Universität Dresden, Heart Centre Dresden, University Hospital, Dresden, Germany
| | - Ruth H Strasser
- Technische Universität Dresden, Heart Centre Dresden, University Hospital, Dresden, Germany
| | - Stephan Wiedemann
- Technische Universität Dresden, Heart Centre Dresden, University Hospital, Dresden, Germany
| |
Collapse
|
48
|
MiR-101 regulates apoptosis of trophoblast HTR-8/SVneo cells by targeting endoplasmic reticulum (ER) protein 44 during preeclampsia. J Hum Hypertens 2014; 28:610-6. [PMID: 24804790 DOI: 10.1038/jhh.2014.35] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/02/2014] [Accepted: 03/26/2014] [Indexed: 12/19/2022]
Abstract
To investigate a possible association between miR-101 and apoptosis of human trophoblast cells mediated by endoplasmic reticulum protein 44 (ERp44) in preeclampsia (PE), we explored the expression of miR-101 in PE placentas (n=30) compared with normotensive pregnant placentas (n=30) and the correlation between miR-101 and ERp44 was also analyzed. Furthermore, both the apoptotic rate of trophoblast cells and the ER stress-induced apoptotic proteins were assayed when the HTR-8/SVneo cells were treated with miR-101 mimics or inhibitors in vitro. We found a lower expression of miR-101 and an inverse correlation between miR-101 and ERp44 protein in PE placentas. Upregulation of miR-101 expression could inhibit trophoblast HTR-8/SVneo cell apoptosis and repress ER stress-induced apoptotic proteins by targeting ERp44 in vitro, whereas inhibition of miR-101 could induce HTR-8/SVneo cell apoptosis. Our findings indicated that overexpression of miR-101 could downregulate ERp44 and suppress apoptosis in trophoblast cells during PE. Therefore, loss of miR-101 expression could contribute to ER stress-induced trophoblast cell apoptosis by targeting ERp44.
Collapse
|
49
|
Piegari E, Lopez L, Perez Ipiña E, Ponce Dawson S. Fluorescence fluctuations and equivalence classes of Ca²⁺ imaging experiments. PLoS One 2014; 9:e95860. [PMID: 24776736 PMCID: PMC4002475 DOI: 10.1371/journal.pone.0095860] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/01/2014] [Indexed: 11/29/2022] Open
Abstract
release into the cytosol through inositol 1,4,5-trisphosphate receptors (IP3Rs) plays a relevant role in numerous physiological processes. IP3R-mediated signals involve -induced -release (CICR) whereby release through one open IP3R induces the opening of other channels. IP3Rs are apparently organized in clusters. The signals can remain localized (i.e., puffs) if CICR is limited to one cluster or become waves that propagate between clusters. puffs are the building blocks of waves. Thus, there is great interest in determining puff properties, especially in view of the current controversy on the spatial distribution of activatable IP3Rs. puffs have been observed in intact cells with optical techniques proving that they are intrinsically stochastic. Obtaining a correct picture of their dynamics then entails being able to detect the whole range of puff sizes. puffs are observed using visible single-wavelength dyes, slow exogenous buffers (e.g., EGTA) to disrupt inter-cluster CICR and UV-photolyzable caged IP3. Single-wavelength dyes increase their fluorescence upon calcium binding producing images that are strongly dependent on their kinetic, transport and photophysical properties. Determining the artifacts that the imaging setting introduces is particularly relevant when trying to analyze the smallest signals. In this paper we introduce a method to estimate the expected signal-to-noise ratio of imaging experiments that use single-wavelength dyes. The method is based on the Number and Brightness technique. It involves the performance of a series of experiments and their subsequent analysis in terms of a fluorescence fluctuation model with which the model parameters are quantified. Using the model, the expected signal-to-noise ratio is then computed. Equivalence classes between different experimental conditions that produce images with similar signal-to-noise ratios can then be established. The method may also be used to estimate the smallest signals that can reliably be observed with each setting.
Collapse
Affiliation(s)
- Estefanía Piegari
- Departamento de Física and IFIBA (CONICET), FCEyN-UBA, Ciudad Universitaria, Pabellón I, Buenos Aires, Argentina
- * E-mail:
| | - Lucía Lopez
- Departamento de Física and IFIBA (CONICET), FCEyN-UBA, Ciudad Universitaria, Pabellón I, Buenos Aires, Argentina
| | - Emiliano Perez Ipiña
- Departamento de Física and IFIBA (CONICET), FCEyN-UBA, Ciudad Universitaria, Pabellón I, Buenos Aires, Argentina
| | - Silvina Ponce Dawson
- Departamento de Física and IFIBA (CONICET), FCEyN-UBA, Ciudad Universitaria, Pabellón I, Buenos Aires, Argentina
| |
Collapse
|
50
|
Akl H, Vervloessem T, Kiviluoto S, Bittremieux M, Parys JB, De Smedt H, Bultynck G. A dual role for the anti-apoptotic Bcl-2 protein in cancer: mitochondria versus endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2240-52. [PMID: 24768714 DOI: 10.1016/j.bbamcr.2014.04.017] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 12/14/2022]
Abstract
Anti-apoptotic Bcl-2 contributes to cancer formation and progression by promoting the survival of altered cells. Hence, it is a prime target for novel specific anti-cancer therapeutics. In addition to its canonical anti-apoptotic role, Bcl-2 has an inhibitory effect on cell-cycle progression. Bcl-2 acts at two different intracellular compartments, the mitochondria and the endoplasmic reticulum (ER). At the mitochondria, Bcl-2 via its hydrophobic cleft scaffolds the Bcl-2-homology (BH) domain 3 (BH3) of pro-apoptotic Bcl-2-family members. Small molecules (like BH3 mimetics) can disrupt this interaction, resulting in apoptotic cell death in cancer cells. At the ER, Bcl-2 modulates Ca(2+) signaling, thereby promoting proliferation while increasing resistance to apoptosis. Bcl-2 at the ER acts via its N-terminal BH4 domain, which directly binds and inhibits the inositol 1,4,5-trisphosphate receptor (IP3R), the main intracellular Ca(2+)-release channel. Tools targeting the BH4 domain of Bcl-2 reverse Bcl-2's inhibitory action on IP3Rs and trigger pro-apoptotic Ca(2+) signaling in cancer B-cells, including chronic lymphocytic leukemia (CLL) cells and diffuse large B-cell lymphoma (DLBCL) cells. The sensitivity of DLBCL cells to BH4-domain targeting tools strongly correlated with the expression levels of the IP3R2 channel, the IP3R isoform with the highest affinity for IP3. Interestingly, bio-informatic analysis of a database of primary CLL patient cells also revealed a transcriptional upregulation of IP3R2. Finally, this review proposes a model, in which cancer cell survival depends on Bcl-2 at the mitochondria and/or the ER. This dependence likely will have an impact on their responses to BH3-mimetic drugs and BH4-domain targeting tools. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.
Collapse
Affiliation(s)
- Haidar Akl
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| | - Tamara Vervloessem
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Santeri Kiviluoto
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Mart Bittremieux
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Humbert De Smedt
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Molecular and Cellular Medicine, Campus Gasthuisberg, O/N-I, Bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|