1
|
Casas-Martinez JC, Samali A, McDonagh B. Redox regulation of UPR signalling and mitochondrial ER contact sites. Cell Mol Life Sci 2024; 81:250. [PMID: 38847861 PMCID: PMC11335286 DOI: 10.1007/s00018-024-05286-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/11/2024] [Accepted: 05/18/2024] [Indexed: 06/13/2024]
Abstract
Mitochondria and the endoplasmic reticulum (ER) have a synergistic relationship and are key regulatory hubs in maintaining cell homeostasis. Communication between these organelles is mediated by mitochondria ER contact sites (MERCS), allowing the exchange of material and information, modulating calcium homeostasis, redox signalling, lipid transfer and the regulation of mitochondrial dynamics. MERCS are dynamic structures that allow cells to respond to changes in the intracellular environment under normal homeostatic conditions, while their assembly/disassembly are affected by pathophysiological conditions such as ageing and disease. Disruption of protein folding in the ER lumen can activate the Unfolded Protein Response (UPR), promoting the remodelling of ER membranes and MERCS formation. The UPR stress receptor kinases PERK and IRE1, are located at or close to MERCS. UPR signalling can be adaptive or maladaptive, depending on whether the disruption in protein folding or ER stress is transient or sustained. Adaptive UPR signalling via MERCS can increase mitochondrial calcium import, metabolism and dynamics, while maladaptive UPR signalling can result in excessive calcium import and activation of apoptotic pathways. Targeting UPR signalling and the assembly of MERCS is an attractive therapeutic approach for a range of age-related conditions such as neurodegeneration and sarcopenia. This review highlights the emerging evidence related to the role of redox mediated UPR activation in orchestrating inter-organelle communication between the ER and mitochondria, and ultimately the determination of cell function and fate.
Collapse
Affiliation(s)
- Jose C Casas-Martinez
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
- Apoptosis Research Centre, University of Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, University of Galway, Galway, Ireland
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland.
- Apoptosis Research Centre, University of Galway, Galway, Ireland.
| |
Collapse
|
2
|
Maleki B, Modarres P, Salehi P, Vallian S. Identification of ITPR1 gene as a novel target for hsa-miR-34b-5p in non-obstructive azoospermia: a Ca 2+/apoptosis pathway cross-talk. Sci Rep 2023; 13:21873. [PMID: 38072953 PMCID: PMC10710998 DOI: 10.1038/s41598-023-49155-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
MiR-34b-5p has been reported as a non-invasive diagnostic biomarker for infertility. However, no gene targets regulating the mechanism of cation of this miRNA are known. In this study, using gene set enrichment analysis the Inositol 1,4,5-Trisphosphate Receptor Type 1 (ITPR1) gene was identified as the sole target for hsa-miR-34b-5p, and found significantly overexpressed in non-obstructive azoospermia (NOA) patients. This finding was confirmed by qRT-PCR on fresh testicular tissues from NOA patients. Then, pathway enrichment analysis as well as the diagnostic value analysis of hsa-miR-34b-5p/ITPR1 indicated ITPR1 as a hub gene in the calcium (Ca2+)-apoptosis pathway, and a valuable predictive biomarker for NOA. Moreover, gene expression and histological assays showed the association of the effects of ITPR1's increased expression on spermatogenesis failure through induction of apoptosis in NOA patients. These data suggested that the hsa-miR-34b-5p/ITPR1 axis could serve as a potential regulatory predictive biomarker for human spermatogenesis through the Ca2+-apoptosis pathway cross-talk.
Collapse
Affiliation(s)
- Bahareh Maleki
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Parastoo Modarres
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Peyman Salehi
- Department of Infertility, Milad Hospital, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Sadeq Vallian
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran.
| |
Collapse
|
3
|
Parthasarathi KTS, Mandal S, George JP, Gaikwad KB, Sasidharan S, Gundimeda S, Jolly MK, Pandey A, Sharma J. Aberrations in ion channels interacting with lipid metabolism and epithelial-mesenchymal transition in esophageal squamous cell carcinoma. Front Mol Biosci 2023; 10:1201459. [PMID: 37529379 PMCID: PMC10388552 DOI: 10.3389/fmolb.2023.1201459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most prevalent malignant gastrointestinal tumor. Ion channels contribute to tumor growth and progression through interactions with their neighboring molecules including lipids. The dysregulation of membrane ion channels and lipid metabolism may contribute to the epithelial-mesenchymal transition (EMT), leading to metastatic progression. Herein, transcriptome profiles of patients with ESCC were analyzed by performing differential gene expression and weighted gene co-expression network analysis to identify the altered ion channels, lipid metabolism- and EMT-related genes in ESCC. A total of 1,081 differentially expressed genes, including 113 ion channels, 487 lipid metabolism-related, and 537 EMT-related genes, were identified in patients with ESCC. Thereafter, EMT scores were correlated with altered co-expressed genes. The altered co-expressed genes indicated a correlation with EMT signatures. Interactions among 22 ion channels with 3 hub lipid metabolism- and 13 hub EMT-related proteins were determined using protein-protein interaction networks. A pathway map was generated to depict deregulated signaling pathways including insulin resistance and the estrogen receptor-Ca2+ signaling pathway in ESCC. The relationship between potential ion channels and 5-year survival rates in ESCC was determined using Kaplan-Meier plots and Cox proportional hazard regression analysis. Inositol 1,4,5-trisphosphate receptor type 3 (ITPR3) was found to be associated with poor prognosis of patients with ESCC. Additionally, drugs interacting with potential ion channels, including GJA1 and ITPR3, were identified. Understanding alterations in ion channels with lipid metabolism and EMT in ESCC pathophysiology would most likely provide potential targets for the better treatment of patients with ESCC.
Collapse
Affiliation(s)
- K. T. Shreya Parthasarathi
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Susmita Mandal
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - John Philip George
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | | | - Sruthi Sasidharan
- Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - Seetaramanjaneyulu Gundimeda
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Mohit Kumar Jolly
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Rochester, MN, United States
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
- Center for Individualized Medicine, Rochester, MN, United States
| | - Jyoti Sharma
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
4
|
Benson JC, Trebak M. Too much of a good thing: The case of SOCE in cellular apoptosis. Cell Calcium 2023; 111:102716. [PMID: 36931194 PMCID: PMC10481469 DOI: 10.1016/j.ceca.2023.102716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
Intracellular calcium (Ca2+) is an essential second messenger in eukaryotic cells regulating numerous cellular functions such as contraction, secretion, immunity, growth, and metabolism. Ca2+ signaling is also a key signal transducer in the intrinsic apoptosis pathway. The store-operated Ca2+ entry pathway (SOCE) is ubiquitously expressed in eukaryotic cells, and is the primary Ca2+ influx pathway in non-excitable cells. SOCE is mediated by the endoplasmic reticulum Ca2+ sensing STIM proteins, and the plasma membrane Ca2+-selective Orai channels. A growing number of studies have implicated SOCE in regulating cell death primarily via the intrinsic apoptotic pathway in a variety of tissues and in response to physiological stressors such as traumatic brain injury, ischemia reperfusion injury, sepsis, and alcohol toxicity. Notably, the literature points to excessive cytosolic Ca2+ influx through SOCE in vulnerable cells as a key factor tipping the balance towards cellular apoptosis. While the literature primarily addresses the functions of STIM1 and Orai1, STIM2, Orai2 and Orai3 are also emerging as potential regulators of cell death. Here, we review the functions of STIM and Orai proteins in regulating cell death and the implications of this regulation to human pathologies.
Collapse
Affiliation(s)
- J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Department of Cellular and Molecular Physiology, Graduate Program, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 1526, USA.
| |
Collapse
|
5
|
Wang Y, Dai G, Lin Z, Cheng C, Zhou X, Song M, Chen P, Ma S, Hu Y, Liu G, Yu B. TWIST1 rescue calcium overload and apoptosis induced by inflammatory microenvironment in S. aureus-induced osteomyelitis. Int Immunopharmacol 2023; 119:110153. [PMID: 37071966 DOI: 10.1016/j.intimp.2023.110153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/13/2023] [Accepted: 04/01/2023] [Indexed: 04/20/2023]
Abstract
Currently, there is no effective therapy for Staphylococcus aureus-induced osteomyelitis. It is widely recognized that the inflammatory microenvironment around abscess plays an essential role in protracting the course of S. aureus-induced osteomyelitis. In this study, we found TWIST1 was highly expressed in macrophages around abscesses but less related to local S. aureus in the later stages of Staphylococcus aureus-infected osteomyelitis. Mouse bone marrow macrophages show apoptosis and elevated TWIST1 expression when treated with the inflammatory medium. Knockdown of TWIST1 induced macrophage apoptosis, impaired the bacteria phagocytosis/killing abilities, and promoted cell apoptosis markers expression in inflammatory microenvironment stimulation. Furthermore, inflammatory microenvironments were responsible for inducing calcium overload in macrophage mitochondrial while calcium overload inhibition significantly rescued macrophage apoptosis, bacteria phagocytosis/killing abilities and improved the mice's antimicrobial ability. Our findings indicated that TWIST1 is a crucial molecule that protects macrophages from calcium overload induced by inflammatory microenvironments.
Collapse
Affiliation(s)
- Yutian Wang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guandong Dai
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zexin Lin
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Caiyu Cheng
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuyou Zhou
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingrui Song
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopedics, Hainan General Hospital (Hainan Affiliated Hospital of Hainan, Medical University), Haikou, China
| | - Sushuang Ma
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Orthopaedics, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Yanjun Hu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanqiao Liu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Bin Yu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Park WY, Gray JM, Holewinski RJ, Andresson T, So JY, Carmona-Rivera C, Hollander MC, Yang HH, Lee M, Kaplan MJ, Cappell SD, Yang L. Apoptosis-induced nuclear expulsion in tumor cells drives S100a4-mediated metastatic outgrowth through the RAGE pathway. NATURE CANCER 2023; 4:419-435. [PMID: 36973439 PMCID: PMC10042736 DOI: 10.1038/s43018-023-00524-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2023] [Indexed: 03/29/2023]
Abstract
Most tumor cells undergo apoptosis in circulation and at the metastatic organ sites due to host immune surveillance and a hostile microenvironment. It remains to be elucidated whether dying tumor cells have a direct effect on live tumor cells during the metastatic process and what the underlying mechanisms are. Here we report that apoptotic cancer cells enhance the metastatic outgrowth of surviving cells through Padi4-mediated nuclear expulsion. Tumor cell nuclear expulsion results in an extracellular DNA-protein complex that is enriched with receptor for advanced glycation endproducts (RAGE) ligands. The chromatin-bound RAGE ligand S100a4 activates RAGE receptors in neighboring surviving tumor cells, leading to Erk activation. In addition, we identified nuclear expulsion products in human patients with breast, bladder and lung cancer and a nuclear expulsion signature correlated with poor prognosis. Collectively, our study demonstrates how apoptotic cell death can enhance the metastatic outgrowth of neighboring live tumor cells.
Collapse
Affiliation(s)
- Woo-Yong Park
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin M Gray
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Ronald J Holewinski
- Protein Mass Spectrometry Group, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Thorkell Andresson
- Protein Mass Spectrometry Group, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Jae Young So
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Christine Hollander
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maxwell Lee
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven D Cappell
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Li Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
High oxygen-modified packaging (HiOx-MAP) mediates HIF-1α regulation of tenderness changes during postmortem aging of yak meat. Food Chem X 2023; 17:100573. [PMID: 36845525 PMCID: PMC9945635 DOI: 10.1016/j.fochx.2023.100573] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/10/2023] Open
Abstract
In the present study, we studied the effect of high oxygen-modified packaging (HiOx-MAP) on yak meat tenderness and the underlying mechanism. HiOx-MAP significantly increased the myofibril fragmentation index (MFI) of yak meat. In addition, western blotting showed that the expression of hypoxia-inducible factor (HIF-1α) and ryanodine receptors (RyR) in the HiOx-MAP group was reduced. HiOx-MAP increased the activity of sarcoplasmic reticulum calcium-ATPase (SERCA). The energy disperse spectroscopy (EDS) mapping showed gradually reduced calcium distribution in the treated endoplasmic reticulum. Furthermore, HiOx-MAP treatment increased the caspase-3 activity and the apoptosis rate. The activity of calmodulin protein (CaMKKβ) and AMP-activated protein kinase (AMPK) was down-regulated leading to apoptosis. These results indicated that HiOx-MAP promoted apoptosis during postmortem aging to improve the tenderization of meat.
Collapse
|
8
|
Rosa N, Speelman-Rooms F, Parys JB, Bultynck G. Modulation of Ca 2+ signaling by antiapoptotic Bcl-2 versus Bcl-xL: From molecular mechanisms to relevance for cancer cell survival. Biochim Biophys Acta Rev Cancer 2022; 1877:188791. [PMID: 36162541 DOI: 10.1016/j.bbcan.2022.188791] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Members of the Bcl-2-protein family are key controllers of apoptotic cell death. The family is divided into antiapoptotic (including Bcl-2 itself, Bcl-xL, Mcl-1, etc.) and proapoptotic members (Bax, Bak, Bim, Bim, Puma, Noxa, Bad, etc.). These proteins are well known for their canonical role in the mitochondria, where they control mitochondrial outer membrane permeabilization and subsequent apoptosis. However, several proteins are recognized as modulators of intracellular Ca2+ signals that originate from the endoplasmic reticulum (ER), the major intracellular Ca2+-storage organelle. More than 25 years ago, Bcl-2, the founding member of the family, was reported to control apoptosis through Ca2+ signaling. Further work elucidated that Bcl-2 directly targets and inhibits inositol 1,4,5-trisphosphate receptors (IP3Rs), thereby suppressing proapoptotic Ca2+ signaling. In addition to Bcl-2, Bcl-xL was also shown to impact cell survival by sensitizing IP3R function, thereby promoting prosurvival oscillatory Ca2+ release. However, new work challenges this model and demonstrates that Bcl-2 and Bcl-xL can both function as inhibitors of IP3Rs. This suggests that, depending on the cell context, Bcl-xL could support very distinct Ca2+ patterns. This not only raises several questions but also opens new possibilities for the treatment of Bcl-xL-dependent cancers. In this review, we will discuss the similarities and divergences between Bcl-2 and Bcl-xL regarding Ca2+ homeostasis and IP3R modulation from both a molecular and a functional point of view, with particular emphasis on cancer cell death resistance mechanisms.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
9
|
Overexpression of the miR-17-92 cluster in colorectal adenoma organoids causes a carcinoma-like gene expression signature. Neoplasia 2022; 32:100820. [PMID: 35872559 PMCID: PMC9307940 DOI: 10.1016/j.neo.2022.100820] [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: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022]
Abstract
Gain of chromosome arm 13q is one of the most prevalent DNA copy number alterations associated with colorectal adenoma-to-carcinoma progression. The oncogenic miR-17-92 cluster, located at 13q, was found to be overexpressed in colorectal cancer and in adenomas harboring 13q gain. However, to what extent overexpression of this group of microRNAs actually drives progression to cancer remains to be resolved. Therefore, we aimed to clarify the role of miR-17-92 cluster in the progression from colorectal adenoma to carcinoma. The miR-17-92 cluster was overexpressed in human colorectal adenoma organoids without 13q gain and downstream effects on mRNA expression were investigated, along with functional consequences in vitro and in vivo. Comparison of mRNA sequencing results of organoids overexpressing miR-17-92 and cultures transduced with control vector revealed a miR-17-92 expression signature. This signature appeared to be enriched in an independent series of colorectal cancers and adenomas with 13q gain, confirming that miR-17-92 expression is associated with malignant progression. However, tumor-associated characteristics, such as increased proliferation rate, were not observed in miR-17-92 overexpressing adenoma organoids in vitro. In addition, subcutaneous injection of these organoids in immunodeficient mice was insufficient to cause tumor outgrowth. In conclusion, this study showed that miR-17-92 expression contributes to 13q gain-associated adenoma-to-carcinoma progression, however, this is insufficient to cause malignancy.
Collapse
|
10
|
Zheng W, Bai X, Zhou Y, Yu L, Ji D, Zheng Y, Meng N, Wang H, Huang Z, Chen W, Yam JWP, Xu Y, Cui Y. Transcriptional ITPR3 as potential targets and biomarkers for human pancreatic cancer. Aging (Albany NY) 2022; 14:4425-4444. [PMID: 35580861 PMCID: PMC9186782 DOI: 10.18632/aging.204080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
Abstract
Inositol 1,4,5-Triphosphate Receptor Family (ITPRs) are necessary intracellular Ca2+-release channel encoders and participate in mammalian cell physiological and pathological processes. Previous studies have suggested that ITPRs participate in tumorigenesis of multiple cancers. Nevertheless, the diverse expression profiles and prognostic significance of three ITPRs in pancreatic cancer have yet to be uncovered. In this work, we examined the expression levels and survival dates of ITPRs in patients with pancreatic cancer. As a result, we identified that ITPR1 and ITPR3 expression levels are significantly elevated in cancerous specimens. Survival data revealed that over-expression of ITPR2 and ITPR3 resulted in unfavourable overall survival and pathological stage. The multivariate Cox logistic regression analysis showed that ITPR3 could be an independent risk factor for PAAD patient survival. Moreover, to investigate how ITPRs work, co-expressed genes, alterations, protein-protein interaction, immune infiltration, methylation, and functional enrichment of ITPRs were also analyzed. Then, we evaluated these findings in clinical samples. Moreover, the gain and loss of function of ITPR3 were also conducted. The electron microscope assay was employed to explore the role of ITPR3 in pancreatic cancer cell lines' endoplasmic reticulum stress. In summary, our findings demonstrated that ITPR3 has the potential to be drug targets and biomarkers for human pancreatic cancer.
Collapse
Affiliation(s)
- Wangyang Zheng
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department II of Gastroenterology, Third Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xue Bai
- Department of Clinic of Internal Medicine I, Ulm University, Ulm 89081, Germany
| | - Yongxu Zhou
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
| | - Liang Yu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
| | - Daolin Ji
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department of Hepatopancreatobiliary Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yuling Zheng
- Department of Pediatric, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Nanfeng Meng
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Hang Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ziyue Huang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wangming Chen
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150086, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
11
|
Inositol (1,4,5)-Trisphosphate Receptors in Invasive Breast Cancer: A New Prognostic Tool? Int J Mol Sci 2022; 23:ijms23062962. [PMID: 35328381 PMCID: PMC8955728 DOI: 10.3390/ijms23062962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The inositol-trisphosphate receptor (IP3R) is a key player in physiological and pathological intracellular calcium signaling. The objective of the present study was to assess the putative value of the three IP3R subtypes as prognostic biomarkers in breast cancer. We found that IP3R3 is the most strongly expressed subtype in breast cancer tissue. Furthermore, IP3R3 and IP3R1 are significantly more expressed in invasive breast cancer tissue than in non-tumor tissue. In contrast to IP3R1 and IP3R2, the expression of IP3R3 was positively correlated with prognostic factors including tumor size, regional node invasion, histologic grade, proliferation index, and hormonal status. By analyzing public databases, we found that the expression of all IP3R subtypes is significantly correlated with the overall survival and disease-free survival of patients with breast cancer. We conclude that relative to the other two IP3R subtypes, IP3R3 expression is upregulated in breast cancer and is correlated with prognostic factors. We strongly believe that our results will open up new perspectives with regard to the link between IP3Rs and breast cancer aggressiveness. Abstract Breast cancer is the leading cause of cancer death among women in worldwide and France. The disease prognosis and treatment differ from one breast cancer subtype to another, and the disease outcome depends on many prognostic factors. Deregulation of ion flux (especially Ca2+ flux) is involved in many pathophysiology processes, including carcinogenesis. Inside the cell, the inositol-trisphosphate receptor (IP3R) is a major player in the regulation of the Ca2+ flux from the endoplasmic reticulum to the cytoplasm. The IP3Rs (and particularly the IP3R3 subtype) are known to be involved in proliferation, migration, and invasion processes in breast cancer cell lines. The objective of the present study was to evaluate the potential value of IP3Rs as prognostic biomarkers in breast cancer. We found that expression levels of IP3R3 and IP3R1 (but not IP3R2) were significantly higher in invasive breast cancer of no special type than in non-tumor tissue from the same patient. However, the IP3R3 subtype was expressed more strongly than the IP3R1 and IP3R2 subtypes. Furthermore, the expression of IP3R3 (but not of IP3R1 or IP3R2) was positively correlated with prognostic factors such as tumor size, regional node invasion, histologic grade, proliferation index, and hormone receptor status. In an analysis of public databases, we found that all IP3Rs types are significantly associated with overall survival and progression-free survival in patients with breast cancer. We conclude that relative to the other two IP3R subtypes, IP3R3 expression is upregulated in breast cancer and is correlated with prognostic factors.
Collapse
|
12
|
Isaacs JT, Brennen WN, Christensen SB, Denmeade SR. Mipsagargin: The Beginning-Not the End-of Thapsigargin Prodrug-Based Cancer Therapeutics. Molecules 2021; 26:7469. [PMID: 34946547 PMCID: PMC8707208 DOI: 10.3390/molecules26247469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/02/2022] Open
Abstract
Søren Brøgger Christensen isolated and characterized the cell-penetrant sesquiterpene lactone Thapsigargin (TG) from the fruit Thapsia garganica. In the late 1980s/early 1990s, TG was supplied to multiple independent and collaborative groups. Using this TG, studies documented with a large variety of mammalian cell types that TG rapidly (i.e., within seconds to a minute) penetrates cells, resulting in an essentially irreversible binding and inhibiting (IC50~10 nM) of SERCA 2b calcium uptake pumps. If exposure to 50-100 nM TG is sustained for >24-48 h, prostate cancer cells undergo apoptotic death. TG-induced death requires changes in the cytoplasmic Ca2+, initiating a calmodulin/calcineurin/calpain-dependent signaling cascade that involves BAD-dependent opening of the mitochondrial permeability transition pore (MPTP); this releases cytochrome C into the cytoplasm, activating caspases and nucleases. Chemically unmodified TG has no therapeutic index and is poorly water soluble. A TG analog, in which the 8-acyl groups is replaced with the 12-aminododecanoyl group, afforded 12-ADT, retaining an EC50 for killing of <100 nM. Conjugation of 12-ADT to a series of 5-8 amino acid peptides was engineered so that they are efficiently hydrolyzed by only one of a series of proteases [e.g., KLK3 (also known as Prostate Specific Antigen); KLK2 (also known as hK2); Fibroblast Activation Protein Protease (FAP); or Folh1 (also known as Prostate Specific Membrane Antigen)]. The obtained conjugates have increased water solubility for systemic delivery in the blood and prevent cell penetrance and, thus, killing until the TG-prodrug is hydrolyzed by the targeting protease in the vicinity of the cancer cells. We summarize the preclinical validation of each of these TG-prodrugs with special attention to the PSMA TG-prodrug, Mipsagargin, which is in phase II clinical testing.
Collapse
Affiliation(s)
- John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - William Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Samuel R. Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (W.N.B.); (S.R.D.)
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
13
|
Calcium Signaling Regulates Autophagy and Apoptosis. Cells 2021; 10:cells10082125. [PMID: 34440894 PMCID: PMC8394685 DOI: 10.3390/cells10082125] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) functions as a second messenger that is critical in regulating fundamental physiological functions such as cell growth/development, cell survival, neuronal development and/or the maintenance of cellular functions. The coordination among various proteins/pumps/Ca2+ channels and Ca2+ storage in various organelles is critical in maintaining cytosolic Ca2+ levels that provide the spatial resolution needed for cellular homeostasis. An important regulatory aspect of Ca2+ homeostasis is a store operated Ca2+ entry (SOCE) mechanism that is activated by the depletion of Ca2+ from internal ER stores and has gained much attention for influencing functions in both excitable and non-excitable cells. Ca2+ has been shown to regulate opposing functions such as autophagy, that promote cell survival; on the other hand, Ca2+ also regulates programmed cell death processes such as apoptosis. The functional significance of the TRP/Orai channels has been elaborately studied; however, information on how they can modulate opposing functions and modulate function in excitable and non-excitable cells is limited. Importantly, perturbations in SOCE have been implicated in a spectrum of pathological neurodegenerative conditions. The critical role of autophagy machinery in the pathogenesis of neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and Huntington’s diseases, would presumably unveil avenues for plausible therapeutic interventions for these diseases. We thus review the role of SOCE-regulated Ca2+ signaling in modulating these diverse functions in stem cell, immune regulation and neuromodulation.
Collapse
|
14
|
Hashemi S, Hosseini SM, Ghalyanchilangeroudi A, Sheikhi N. Transcriptome based analysis of apoptosis genes in chickens co-infected with avian infectious bronchitis virus and pathogenic Escherichia coli. IRANIAN JOURNAL OF MICROBIOLOGY 2021; 13:17-22. [PMID: 33889358 PMCID: PMC8043830 DOI: 10.18502/ijm.v13i1.5487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVES Infection with Infectious bronchitis virus (IBV) and avian pathogenic Escherichia coli (APEC) is an important respiratory infection worldwide. Apoptosis is a physiological process of cell death that occurs as part of normal development and responds to a variety of physiological and pathophysiological stimuli. The identification of molecular mechanisms of action or inaction of key apoptotic proteins is important. This study aimed to investigate apoptotic related genes in the trachea tissue of infected (IBV variant 2, and APEC serotype O78: K80) SPF chickens group compared to the control group. MATERIALS AND METHODS Forty SPF chickens was divided into 2 groups. Differential transcriptional profile in the infected SPF chickens trachea tissue was compared to those of control group in the early stage of infection by Illumina RNA-seq technique paired-end and strand-specific sequencing. Differentially expressed genes (DEGs) of transcriptome profiling of the trachea from the infected group were identified. Gene ontology category, KEGG pathway, and STRING analysis were analyzed to identify relationships among differentially expressed genes. RESULTS Twenty-eight apoptotic genes were identified. They consisted of six pathways related to cell death: the extrinsic pathway, intrinsic pathway, endoplasmic reticulum stress pathway, MAPK signaling pathway, and cell death by NFkB and activates mTOR pathway and some regulator and apoptosis inhibitors. CONCLUSION All of the apoptotic genes in our study were up-regulated. Among these genes, the more fold change value was for TRADD and BCL2A1 genes, and the less fold change value was for MAP3K14, NFKB1, PIK3CB, and ITPR2 genes.
Collapse
Affiliation(s)
- Shabnam Hashemi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Seyed Masoud Hosseini
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Arash Ghalyanchilangeroudi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Nariman Sheikhi
- Department of Clinical Sciences, College of Veterinary, Tehran Sciences and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
15
|
Gambardella J, Morelli MB, Wang X, Castellanos V, Mone P, Santulli G. The discovery and development of IP3 receptor modulators: an update. Expert Opin Drug Discov 2021; 16:709-718. [PMID: 33356639 DOI: 10.1080/17460441.2021.1858792] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Introduction: Inositol 1,4,5-trisphosphate receptors (IP3Rs) are intracellular calcium (Ca2+) release channels located on the endoplasmic/sarcoplasmic reticulum. The availability of the structure of the ligand-binding domain of IP3Rs has enabled the design of compatible ligands, but the limiting step remains their actual effectiveness in a biological context.Areas covered: This article summarizes the compelling literature on both agonists and antagonists targeting IP3Rs, emphasizing their strengths and limitations. The main challenges toward the discovery and development of IP3 receptor modulators are also described.Expert opinion: Despite significant progress in recent years, the pharmacology of IP3R still has major drawbacks, especially concerning the availability of specific antag onists. Moreover, drugs specifically targeting the three different subtypes of IP3R are especially needed.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York City, USA.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, USA.,Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.,International Translational Research and Medical Education (ITME), Naples, Italy
| | - Marco B Morelli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York City, USA.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, USA
| | - Xujun Wang
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York City, USA
| | - Vanessa Castellanos
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York City, USA
| | - Pasquale Mone
- University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, Montefiore University Hospital, New York City, USA.,Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Einstein-Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, USA.,Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy.,International Translational Research and Medical Education (ITME), Naples, Italy
| |
Collapse
|
16
|
Rosa N, Sneyers F, Parys JB, Bultynck G. Type 3 IP 3 receptors: The chameleon in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:101-148. [PMID: 32247578 DOI: 10.1016/bs.ircmb.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), intracellular calcium (Ca2+) release channels, fulfill key functions in cell death and survival processes, whose dysregulation contributes to oncogenesis. This is essentially due to the presence of IP3Rs in microdomains of the endoplasmic reticulum (ER) in close proximity to the mitochondria. As such, IP3Rs enable efficient Ca2+ transfers from the ER to the mitochondria, thus regulating metabolism and cell fate. This review focuses on one of the three IP3R isoforms, the type 3 IP3R (IP3R3), which is linked to proapoptotic ER-mitochondrial Ca2+ transfers. Alterations in IP3R3 expression have been highlighted in numerous cancer types, leading to dysregulations of Ca2+ signaling and cellular functions. However, the outcome of IP3R3-mediated Ca2+ transfers for mitochondrial function is complex with opposing effects on oncogenesis. IP3R3 can either suppress cancer by promoting cell death and cellular senescence or support cancer by driving metabolism, anabolic processes, cell cycle progression, proliferation and invasion. The aim of this review is to provide an overview of IP3R3 dysregulations in cancer and describe how such dysregulations alter critical cellular processes such as proliferation or cell death and survival. Here, we pose that the IP3R3 isoform is not only linked to proapoptotic ER-mitochondrial Ca2+ transfers but might also be involved in prosurvival signaling.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium.
| |
Collapse
|
17
|
Type 3 inositol 1,4,5-trisphosphate receptor: A calcium channel for all seasons. Cell Calcium 2019; 85:102132. [PMID: 31790953 DOI: 10.1016/j.ceca.2019.102132] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Inositol 1,4,5 trisphosphate receptors (ITPRs) are a family of endoplasmic reticulum Ca2+ channels essential for the control of intracellular Ca2+ levels in virtually every mammalian cell type. The three isoforms (ITPR1, ITPR2 and ITPR3) are highly homologous in amino acid sequence, but they differ considerably in terms of biophysical properties, subcellular localization, and tissue distribution. Such differences underscore the variety of cellular responses triggered by each isoform and suggest that the expression/activity of specific isoforms might be linked to particular pathophysiological states. Indeed, recent findings demonstrate that changes in expression of ITPR isoforms are associated with a number of human diseases ranging from fatty liver disease to cancer. ITPR3 is emerging as the isoform that is particularly important in the pathogenesis of various human diseases. Here we review the physiological and pathophysiological roles of ITPR3 in various tissues and the mechanisms by which the expression of this isoform is modulated in health and disease.
Collapse
|
18
|
Abstract
In the body, extracellular stimuli produce inositol 1,4,5-trisphosphate (IP3), an intracellular chemical signal that binds to the IP3 receptor (IP3R) to release calcium ions (Ca2+) from the endoplasmic reticulum. In the past 40 years, the wide-ranging functions mediated by IP3R and its genetic defects causing a variety of disorders have been unveiled. Recent cryo-electron microscopy and X-ray crystallography have resolved IP3R structures and begun to integrate with concurrent functional studies, which can explicate IP3-dependent opening of Ca2+-conducting gates placed ∼90 Å away from IP3-binding sites and its regulation by Ca2+. This review highlights recent research progress on the IP3R structure and function. We also propose how protein plasticity within IP3R, which involves allosteric gating and assembly transformations accompanied by rapid and chronic structural changes, would enable it to regulate diverse functions at cellular microdomains in pathophysiological states.
Collapse
Affiliation(s)
- Kozo Hamada
- Laboratory of Cell Calcium Signaling, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China; ,
| | - Katsuhiko Mikoshiba
- Laboratory of Cell Calcium Signaling, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China; ,
| |
Collapse
|
19
|
Carbone M, Amelio I, Affar EB, Brugarolas J, Cannon-Albright LA, Cantley LC, Cavenee WK, Chen Z, Croce CM, Andrea AD, Gandara D, Giorgi C, Jia W, Lan Q, Mak TW, Manley JL, Mikoshiba K, Onuchic JN, Pass HI, Pinton P, Prives C, Rothman N, Sebti SM, Turkson J, Wu X, Yang H, Yu H, Melino G. Consensus report of the 8 and 9th Weinman Symposia on Gene x Environment Interaction in carcinogenesis: novel opportunities for precision medicine. Cell Death Differ 2018; 25:1885-1904. [PMID: 30323273 PMCID: PMC6219489 DOI: 10.1038/s41418-018-0213-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The relative contribution of intrinsic genetic factors and extrinsic environmental ones to cancer aetiology and natural history is a lengthy and debated issue. Gene-environment interactions (G x E) arise when the combined presence of both a germline genetic variant and a known environmental factor modulates the risk of disease more than either one alone. A panel of experts discussed our current understanding of cancer aetiology, known examples of G × E interactions in cancer, and the expanded concept of G × E interactions to include somatic cancer mutations and iatrogenic environmental factors such as anti-cancer treatment. Specific genetic polymorphisms and genetic mutations increase susceptibility to certain carcinogens and may be targeted in the near future for prevention and treatment of cancer patients with novel molecularly based therapies. There was general consensus that a better understanding of the complexity and numerosity of G × E interactions, supported by adequate technological, epidemiological, modelling and statistical resources, will further promote our understanding of cancer and lead to novel preventive and therapeutic approaches.
Collapse
Affiliation(s)
| | | | - El Bachir Affar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, Quebec, H1T 2M4, Canada
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY, 10021, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhijian Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alan D' Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David Gandara
- Thoracic Oncology, UC Davis, Sacramento, CA, 96817, USA
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Wei Jia
- Hawaii Cancer Center, Honolulu, HI, USA
| | - Qing Lan
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Tak Wah Mak
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Jose N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
| | - Harvey I Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, 10027, USA
| | - Nathaniel Rothman
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Said M Sebti
- Drug Discovery Department, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida, Tampa, FL, 33612, USA
| | | | - Xifeng Wu
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Gerry Melino
- MRC Toxicology Unit, Leicester, UK.
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
20
|
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: 4.2] [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
|
21
|
ITPR3 gene haplotype is associated with cervical squamous cell carcinoma risk in Taiwanese women. Oncotarget 2018; 8:10085-10090. [PMID: 28036301 PMCID: PMC5354643 DOI: 10.18632/oncotarget.14341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/15/2016] [Indexed: 11/25/2022] Open
Abstract
Host immunogenetic background plays an important role in human papillomavirus (HPV) infection and cervical cancer development. Inositol 1,4,5-triphosphate receptor type 3 (ITPR3) is essential for both immune activation and cancer pathogenesis. We aim to investigate if ITPR3 genetic polymorphisms are associated with the risk of cervical cancer in Taiwanese women. ITPR3 rs3748079 A/G and rs2229634 C/T polymorphisms were genotyped in a hospital-based study of 462 women with cervical squamous cell carcinoma (CSCC) and 921 age-matched healthy control women. The presence and genotypes of HPV in CSCC was determined. No significant association of individual ITPR3 variants were found among controls, CSCC, and HPV-16 positive CSCC. However, we found a significant association of haplotype AT between CSCC and controls (OR = 2.28, 95% CI 1.31-3.97, P = 2.83 × 10-3) and the OR increased further in CSCC patients infected with HPV-16 (OR = 2.89, 95% CI 1.55-5.37, P = 4.54 × 10-4). The linkage disequilibrium analysis demonstrated that ITPR3 association with CSCC was independent of HLA-DRB1 alleles. In conclusion, these findings suggest that AT haplotype in the ITPR3 gene may serve as a potential marker for genetic susceptibility to CSCC.
Collapse
|
22
|
Bijnens J, Missiaen L, Bultynck G, Parys JB. A critical appraisal of the role of intracellular Ca 2+-signaling pathways in Kawasaki disease. Cell Calcium 2018; 71:95-103. [PMID: 29604968 DOI: 10.1016/j.ceca.2018.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/20/2018] [Indexed: 12/31/2022]
Abstract
Kawasaki disease is a multi-systemic vasculitis that generally occurs in children and that can lead to coronary artery lesions. Recent studies showed that Kawasaki disease has an important genetic component. In this review, we discuss the single-nucleotide polymorphisms in the genes encoding proteins with a role in intracellular Ca2+ signaling: inositol 1,4,5-trisphosphate 3-kinase C, caspase-3, the store-operated Ca2+-entry channel ORAI1, the type-3 inositol 1,4,5-trisphosphate receptor, the Na+/Ca2+ exchanger 1, and phospholipase Cß4 and Cß1. An increase of the free cytosolic Ca2+ concentration is proposed to be a major factor in susceptibility to Kawasaki disease and disease outcome, but only for polymorphisms in the genes encoding the inositol 1,4,5-trisphosphate 3-kinase C and the Na+/Ca2+ exchanger 1, the free cytosolic Ca2+ concentration was actually measured and shown to be increased. Excessive cytosolic Ca2+ signaling can result in hyperactive calcineurin in T cells with an overstimulated nuclear factor of activated T cells pathway, in hypersecretion of interleukin-1ß and tumor necrosis factor-α by monocytes/macrophages, in increased urotensin-2 signaling, and in an overactivation of vascular endothelial cells.
Collapse
Affiliation(s)
- Jeroen Bijnens
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, B-3000 Leuven, Belgium
| | - Ludwig Missiaen
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, B-3000 Leuven, Belgium.
| |
Collapse
|
23
|
Ando H, Kawaai K, Bonneau B, Mikoshiba K. Remodeling of Ca 2+ signaling in cancer: Regulation of inositol 1,4,5-trisphosphate receptors through oncogenes and tumor suppressors. Adv Biol Regul 2017; 68:64-76. [PMID: 29287955 DOI: 10.1016/j.jbior.2017.12.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022]
Abstract
The calcium ion (Ca2+) is a ubiquitous intracellular signaling molecule that regulates diverse physiological and pathological processes, including cancer. Increasing evidence indicates that oncogenes and tumor suppressors regulate the Ca2+ transport systems. Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) are IP3-activated Ca2+ release channels located on the endoplasmic reticulum (ER). They play pivotal roles in the regulation of cell death and survival by controlling Ca2+ transfer from the ER to mitochondria through mitochondria-associated ER membranes (MAMs). Optimal levels of Ca2+ mobilization to mitochondria are necessary for mitochondrial bioenergetics, whereas excessive Ca2+ flux into mitochondria causes loss of mitochondrial membrane integrity and apoptotic cell death. In addition to well-known functions on outer mitochondrial membranes, B-cell lymphoma 2 (Bcl-2) family proteins are localized on the ER and regulate IP3Rs to control Ca2+ transfer into mitochondria. Another regulatory protein of IP3R, IP3R-binding protein released with IP3 (IRBIT), cooperates with or counteracts the Bcl-2 family member depending on cellular states. Furthermore, several oncogenes and tumor suppressors, including Akt, K-Ras, phosphatase and tensin homolog (PTEN), promyelocytic leukemia protein (PML), BRCA1, and BRCA1 associated protein 1 (BAP1), are localized on the ER or at MAMs and negatively or positively regulate apoptotic cell death through interactions with IP3Rs and regulation of Ca2+ dynamics. The remodeling of Ca2+ signaling by oncogenes and tumor suppressors that interact with IP3Rs has fundamental roles in the pathology of cancers.
Collapse
Affiliation(s)
- Hideaki Ando
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Katsuhiro Kawaai
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Benjamin Bonneau
- Institute NeuroMyoGene (INMG), CNRS UMR 5310, INSERM U1217, Gregor Mendel building, 16, rue Raphaël Dubois, 69100 Villeurbanne, France
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
24
|
Portelli LA, Falldorf K, Thuróczy G, Cuppen J. Retrospective estimation of the electric and magnetic field exposure conditions in in vitro experimental reports reveal considerable potential for uncertainty. Bioelectromagnetics 2017; 39:231-243. [PMID: 29171034 DOI: 10.1002/bem.22099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 10/11/2017] [Indexed: 12/13/2022]
Abstract
Experiments on cell cultures exposed to extremely low frequency (ELF, 3-300 Hz) magnetic fields are often subject to multiple sources of uncertainty associated with specific electric and magnetic field exposure conditions. Here we systemically quantify these uncertainties based on exposure conditions described in a group of bioelectromagnetic experimental reports for a representative sampling of the existing literature. The resulting uncertainties, stemming from insufficient, ambiguous, or erroneous description, design, implementation, or validation of the experimental methods and systems, were often substantial enough to potentially make any successful reproduction of the original experimental conditions difficult or impossible. Without making any assumption about the true biological relevance of ELF electric and magnetic fields, these findings suggest another contributing factor which may add to the overall variability and irreproducibility traditionally associated with experimental results of in vitro exposures to low-level ELF magnetic fields. Bioelectromagnetics. 39:231-243, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lucas A Portelli
- Department of Electrical, Computer and Energy Engineering, University of Colorado, Boulder, Colorado.,The Foundation for Research on Information Technologies in Society (IT'IS), Zurich, Switzerland
| | | | - György Thuróczy
- National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Jan Cuppen
- Immunent B.V. and Neiding B.V., Veldhoven, The Netherlands
| |
Collapse
|
25
|
Genetically encoded calcium indicators for studying long-term calcium dynamics during apoptosis. Cell Calcium 2017; 61:44-49. [PMID: 28073595 DOI: 10.1016/j.ceca.2016.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/31/2016] [Accepted: 12/31/2016] [Indexed: 12/18/2022]
Abstract
Intracellular calcium release is essential for regulating almost all cellular functions. Specific spatio-temporal patterns of cytosolic calcium elevations are critical determinants of cell fate in response to pro-apoptotic cellular stressors. As the apoptotic program can take hours or days, measurement of long-term calcium dynamics are essential for understanding the mechanistic role of calcium in apoptotic cell death. Due to the technical limitations of using calcium-sensitive dyes to measure cytosolic calcium little is known about long-term calcium dynamics in living cells after treatment with apoptosis-inducing drugs. Genetically encoded calcium indicators could potentially overcome some of the limitations of calcium-sensitive dyes. Here, we compared the performance of the genetically encoded calcium indicators GCaMP6s and GCaMP6f with the ratiometric dye Fura-2. GCaMP6s performed as well or better than Fura-2 in detecting agonist-induced calcium transients. We then examined the utility of GCaMP6s for continuously measuring apoptotic calcium release over the course of ten hours after treatment with staurosporine. We found that GCaMP6s was suitable for measuring apoptotic calcium release over long time courses and revealed significant heterogeneity in calcium release dynamics in individual cells challenged with staurosporine. Our results suggest GCaMP6s is an excellent indicator for monitoring long-term changes cytosolic calcium during apoptosis.
Collapse
|
26
|
Chae Y, Kim D, An YJ. Effect of fluoride on the cell viability, cell organelle potential, and photosynthetic capacity of freshwater and soil algae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 219:359-367. [PMID: 27814553 DOI: 10.1016/j.envpol.2016.10.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/18/2016] [Accepted: 10/21/2016] [Indexed: 06/06/2023]
Abstract
Although fluoride occurs naturally in the environment, excessive amounts of fluoride in freshwater and terrestrial ecosystems can be harmful. We evaluated the toxicity of fluoride compounds on the growth, viability, and photosynthetic capacity of freshwater (Chlamydomonas reinhardtii and Pseudokirchneriella subcapitata) and terrestrial (Chlorococcum infusionum) algae. To measure algal growth inhibition, a flow cytometric method was adopted (i.e., cell size, granularity, and auto-fluorescence measurements), and algal yield was calculated to assess cell viability. Rhodamine123 and fluorescein diacetate were used to evaluate mitochondrial membrane potential (MMA, ΔΨm) and cell permeability. Nine parameters related to the photosynthetic capacity of algae were also evaluated. The results indicated that high concentrations of fluoride compounds affected cell viability, cell organelle potential, and photosynthetic functions. The cell viability measurements of the three algal species decreased, but apoptosis was only observed in C. infusionum. The MMA (ΔΨm) of cells exposed to fluoride varied among species, and the cell permeability of the three species generally decreased. The decrease in the photosynthetic activity of algae may be attributable to the combination of fluoride ions (F-) with magnesium ions (Mg2+) in chlorophyll. Our results therefore provide strong evidence for the potential risks of fluoride compounds to microflora and microfauna in freshwater and terrestrial ecosystems.
Collapse
Affiliation(s)
- Yooeun Chae
- Department of Environmental Health Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dokyung Kim
- Department of Environmental Health Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Youn-Joo An
- Department of Environmental Health Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
27
|
Golbach LA, Portelli LA, Savelkoul HFJ, Terwel SR, Kuster N, de Vries RBM, Verburg-van Kemenade BML. Calcium homeostasis and low-frequency magnetic and electric field exposure: A systematic review and meta-analysis of in vitro studies. ENVIRONMENT INTERNATIONAL 2016; 92-93:695-706. [PMID: 26872872 DOI: 10.1016/j.envint.2016.01.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 12/10/2015] [Accepted: 01/18/2016] [Indexed: 05/24/2023]
Abstract
Low frequency magnetic field (LF MF) exposure is recurrently suggested to have the ability to induce health effects in society. Therefore, in vitro model systems are used to investigate biological effects of exposure. LF MF induced changes of the cellular calcium homeostasis are frequently hypothesised to be the possible target, but this hypothesis is both substantiated and rejected by numerous studies in literature. Despite the large amount of data, no systematic analysis of in vitro studies has been conducted to address the strength of evidence for an association between LF MF exposure and calcium homeostasis. Our systematic review, with inclusion of 42 studies, showed evidence for an association of LF MF with internal calcium concentrations and calcium oscillation patterns. The oscillation frequency increased, while the amplitude and the percentage of oscillating cells remained constant. The intracellular calcium concentration increased (SMD 0.351, 95% CI 0.126, 0.576). Subgroup analysis revealed heterogeneous effects associated with the exposure frequency, magnetic flux density and duration. Moreover, we found support for the presence of MF-sensitive cell types. Nevertheless, some of the included studies may introduce a great risk of bias as a result of uncontrolled or not reported exposure conditions, temperature ranges and ambient fields. In addition, mathematical calculations of the parasitic induced electric fields (IEFs) disclosed their association with increased intracellular calcium. Our results demonstrate that LF MF might influence the calcium homeostasis in cells in vitro, but the risk of bias and high heterogeneity (I(2)>75%) weakens the analyses. Therefore any potential clinical implications await further investigation.
Collapse
Affiliation(s)
- Lieke A Golbach
- Cell Biology and Immunology Group, Wageningen University, P.O.Box 338, 6700AH Wageningen, The Netherlands
| | - Lucas A Portelli
- The Foundation for Research on Information Technologies in Society (IT'IS), Zeughausstrasse 43, CH-8004 Zurich, Switzerland
| | - Huub F J Savelkoul
- Cell Biology and Immunology Group, Wageningen University, P.O.Box 338, 6700AH Wageningen, The Netherlands
| | - Sofie R Terwel
- Cell Biology and Immunology Group, Wageningen University, P.O.Box 338, 6700AH Wageningen, The Netherlands
| | - Niels Kuster
- The Foundation for Research on Information Technologies in Society (IT'IS), Zeughausstrasse 43, CH-8004 Zurich, Switzerland; Department of Information Technology and Electrical Engineering, Swiss Federal Technical Institute (ETHZ), Gloriastrasse 35, CH-8092 Zurich, Switzerland
| | - Rob B M de Vries
- SYRCLE, Central Animal Laboratory, Radboud University Medical Center, The Netherlands
| | | |
Collapse
|
28
|
Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N. Ion channels in the regulation of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2532-46. [PMID: 25450339 DOI: 10.1016/j.bbamem.2014.10.030] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/08/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Abstract
Apoptosis, a type of genetically controlled cell death, is a fundamental cellular mechanism utilized by multicellular organisms for disposal of cells that are no longer needed or potentially detrimental. Given the crucial role of apoptosis in physiology, deregulation of apoptotic machinery is associated with various diseases as well as abnormalities in development. Acquired resistance to apoptosis represents the common feature of most and perhaps all types of cancer. Therefore, repairing and reactivating apoptosis represents a promising strategy to fight cancer. Accumulated evidence identifies ion channels as essential regulators of apoptosis. However, the contribution of specific ion channels to apoptosis varies greatly depending on cell type, ion channel type and intracellular localization, pathology as well as intracellular signaling pathways involved. Here we discuss the involvement of major types of ion channels in apoptosis regulation. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France.
| |
Collapse
|
29
|
Wiel C, Lallet-Daher H, Gitenay D, Gras B, Le Calvé B, Augert A, Ferrand M, Prevarskaya N, Simonnet H, Vindrieux D, Bernard D. Endoplasmic reticulum calcium release through ITPR2 channels leads to mitochondrial calcium accumulation and senescence. Nat Commun 2014; 5:3792. [DOI: 10.1038/ncomms4792] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/03/2014] [Indexed: 12/21/2022] Open
|
30
|
Pierro C, Cook SJ, Foets TCF, Bootman MD, Roderick HL. Oncogenic K-Ras suppresses IP₃-dependent Ca²⁺ release through remodelling of the isoform composition of IP₃Rs and ER luminal Ca²⁺ levels in colorectal cancer cell lines. J Cell Sci 2014; 127:1607-19. [PMID: 24522186 DOI: 10.1242/jcs.141408] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The GTPase Ras is a molecular switch engaged downstream of G-protein-coupled receptors and receptor tyrosine kinases that controls multiple cell-fate-determining signalling pathways. Ras signalling is frequently deregulated in cancer, underlying associated changes in cell phenotype. Although Ca(2+) signalling pathways control some overlapping functions with Ras, and altered Ca(2+) signalling pathways are emerging as important players in oncogenic transformation, how Ca(2+) signalling is remodelled during transformation and whether it has a causal role remains unclear. We have investigated Ca(2+) signalling in two human colorectal cancer cell lines and their isogenic derivatives in which the allele encoding oncogenic K-Ras (G13D) was deleted by homologous recombination. We show that agonist-induced Ca(2+) release from the endoplasmic reticulum (ER) intracellular Ca(2+) stores is enhanced by loss of K-Ras(G13D) through an increase in the Ca(2+) content of the ER store and a modification of the abundance of inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) subtypes. Consistently, uptake of Ca(2+) into mitochondria and sensitivity to apoptosis was enhanced as a result of K-Ras(G13D) loss. These results suggest that suppression of Ca(2+) signalling is a common response to naturally occurring levels of K-Ras(G13D), and that this contributes to a survival advantage during oncogenic transformation.
Collapse
Affiliation(s)
- Cristina Pierro
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT UK
| | | | | | | | | |
Collapse
|
31
|
Hsieh YF, Liu GY, Lee YJ, Yang JJ, Sándor K, Sarang Z, Bononi A, Pinton P, Tretter L, Szondy Z, Tsay GJ. Transglutaminase 2 contributes to apoptosis induction in Jurkat T cells by modulating Ca2+ homeostasis via cross-linking RAP1GDS1. PLoS One 2013; 8:e81516. [PMID: 24349085 PMCID: PMC3859493 DOI: 10.1371/journal.pone.0081516] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/23/2013] [Indexed: 12/22/2022] Open
Abstract
Background Transglutaminase 2 (TG2) is a protein cross-linking enzyme known to be associated with the in vivo apoptosis program of T cells. However, its role in the T cell apoptosis program was not investigated yet. Results Here we report that timed overexpression of both the wild type (wt) and the cross-linking mutant of TG2 induced apoptosis in Jurkat T cells, the wt being more effective. Part of TG2 colocalised with mitochondria. WtTG2-induced apoptosis was characterized by enhanced mitochondrial Ca2+ uptake. Ca2+-activated wtTG2 cross-linked RAP1, GTP-GDP dissociation stimulator 1, an unusual guanine exchange factor acting on various small GTPases, to induce a yet uncharacterized signaling pathway that was able to promote the Ca2+ release from the endoplasmic reticulum via both Ins3P and ryanodine sensitive receptors leading to a consequently enhanced mitochondrial Ca2+uptake. Conclusions Our data indicate that TG2 might act as a Ca2+ sensor to amplify endoplasmic reticulum-derived Ca2+ signals to enhance mitochondria Ca2+ uptake. Since enhanced mitochondrial Ca2+ levels were previously shown to sensitize mitochondria for various apoptotic signals, our data demonstrate a novel mechanism through which TG2 can contribute to the induction of apoptosis in certain cell types. Since, as compared to knock out cells, physiological levels of TG2 affected Ca2+ signals in mouse embryonic fibroblasts similar to Jurkat cells, our data might indicate a more general role of TG2 in the regulation of mitochondrial Ca2+ homeostasis.
Collapse
Affiliation(s)
- Yu-Fan Hsieh
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Guang-Yaw Liu
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Ju Lee
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jiann-Jou Yang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Katalin Sándor
- Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Angela Bononi
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Experimental and Diagnostic Medicine, Section of General Pathology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - László Tretter
- Department of Medical Biochemistry, Semmelweis University, Neurobiochemical Group of Hungarian Academy of Sciences, Budapest, Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology, Research Center of Molecular Medicine, University of Debrecen, Debrecen, Hungary
- * E-mail: (ZS); (GJT)
| | - Gregory J. Tsay
- Institute of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail: (ZS); (GJT)
| |
Collapse
|
32
|
Suwanjang W, Abramov AY, Govitrapong P, Chetsawang B. Melatonin attenuates dexamethasone toxicity-induced oxidative stress, calpain and caspase activation in human neuroblastoma SH-SY5Y cells. J Steroid Biochem Mol Biol 2013; 138:116-22. [PMID: 23688838 DOI: 10.1016/j.jsbmb.2013.04.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/11/2013] [Accepted: 04/15/2013] [Indexed: 02/02/2023]
Abstract
Glucocorticoids (GCs) have a significant role in the adaptive response of the brain to stress. Increasing evidence has demonstrated that an increase of GC levels may induce neuronal cell death via apoptotic pathways. There is a correlation between over-production of reactive oxygen species (ROS) and an elevation in cytosolic calcium that causes a subsequent increase in the calcium-dependent death-process activation in GC-induced toxicity. Consequently, melatonin, via its antioxidant activity, exhibits a neuroprotective effect against apoptosis induced by intracellular calcium overload. Therefore, in the present study, we explored the protective effect of melatonin in GC-induced toxicity in dopaminergic SH-SY5Y cells. Cellular treatment with the synthetic GCs, dexamethasone (DEX), resulted in a marked decrease in cell viability and in the level of the calpain-inhibitor protein, calpastatin. DEX-induced toxicity also caused an increase in ROS production and the activation of the calcium-dependent cysteine protease, calpain, along with an increase in caspase-3 activation. Pretreatment of the cells with melatonin substantially prevented the decrease in cell viability, over-production of ROS and the activation of calpain and caspase-3, and reversed the depletion in calpastatin levels. These results suggest that melatonin may exert its protective effects against the calpain- and caspase-dependent death process in DEX-induced neurotoxicity.
Collapse
Affiliation(s)
- Wilasinee Suwanjang
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, Thailand
| | | | | | | |
Collapse
|
33
|
English AR, Voeltz GK. Endoplasmic reticulum structure and interconnections with other organelles. Cold Spring Harb Perspect Biol 2013; 5:a013227. [PMID: 23545422 DOI: 10.1101/cshperspect.a013227] [Citation(s) in RCA: 231] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The endoplasmic reticulum (ER) is a large, continuous membrane-bound organelle comprised of functionally and structurally distinct domains including the nuclear envelope, peripheral tubular ER, peripheral cisternae, and numerous membrane contact sites at the plasma membrane, mitochondria, Golgi, endosomes, and peroxisomes. These domains are required for multiple cellular processes, including synthesis of proteins and lipids, calcium level regulation, and exchange of macromolecules with various organelles at ER-membrane contact sites. The ER maintains its unique overall structure regardless of dynamics or transfer at ER-organelle contacts. In this review, we describe the numerous factors that contribute to the structure of the ER.
Collapse
Affiliation(s)
- Amber R English
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | | |
Collapse
|
34
|
Tespa1 is a novel component of mitochondria-associated endoplasmic reticulum membranes and affects mitochondrial calcium flux. Biochem Biophys Res Commun 2013; 433:322-6. [DOI: 10.1016/j.bbrc.2013.02.099] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 02/28/2013] [Indexed: 12/20/2022]
|
35
|
Fujimoto T, Matsuzaki H, Tanaka M, Shirasawa S. Tespa1 protein is phosphorylated in response to store-operated calcium entry. Biochem Biophys Res Commun 2013; 434:162-5. [PMID: 23541577 DOI: 10.1016/j.bbrc.2013.02.128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
We previously reported that Tespa1 (thymocyte-expressed, positive selection-associated gene 1) protein expressed in lymphocytes physically interacts with IP3R (Inositol 1,4,5-trisphosphate receptor), a Ca(2+) channel protein spanning endoplasmic reticulum (ER) membrane. However, the biochemical characterization of Tespa1 protein remains unknown. In this study, we have found that Tespa1 protein was posttranslationally modified upon intracellular Ca(2+) increase in thymocytes. Through the analyses using various inhibitors, store-operated Ca(2+) entry (SOCE) was found to be an essential factor for the Tespa1 protein modification induced by T cell receptor (TCR)-stimulation. Remarkably, the Ca(2+)-dependent Tespa1 protein modification was restored by in vitro protein phosphatase treatment, indicating that this modification was due to phosphorylation. Moreover, we examined whether Ca(2+)-dependent phosphorylation of Tespa1 protein would affect the physical association between Tespa1 and IP3R proteins, revealing that physical association of these proteins is maintained regardless of the presence or absence of phosphorylation of Tespa1. In addition, KRAP protein which represents substantial amino acid sequence homology to Tespa1 was also posttranslationally phosphorylated by intracellular Ca(2+) increase in HCT116 human colon cancer cells and HEK293 human embryonic kidney cells, suggesting that common signaling mechanism(s) may contribute to the molecular modification of Tespa1 and KRAP in different cellular processes. All these results suggested a novel molecular modification of Tespa1 and the existence of the regulatory pathway that SOCE affects the Tespa1-IP3R molecular complex.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Fukuoka 814-0180, Japan
| | | | | | | |
Collapse
|
36
|
Alzayady KJ, Chandrasekhar R, Yule DI. Fragmented inositol 1,4,5-trisphosphate receptors retain tetrameric architecture and form functional Ca2+ release channels. J Biol Chem 2013; 288:11122-34. [PMID: 23479737 DOI: 10.1074/jbc.m113.453241] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptor isoforms are a family of ubiquitously expressed ligand-gated channels encoded by three individual genes. The proteins are localized to membranes of intracellular Ca(2+) stores and play pivotal roles in Ca(2+) homeostasis. Previous studies have demonstrated that IP3R1 is cleaved by the intracellular proteases calpain and caspase both in vivo and in vitro. However, the resultant cleavage products are poorly defined, and the functional consequences of these proteolytic events are not fully understood. We demonstrate that IP3R1 is cleaved during staurosporine-induced apoptosis, yielding N-terminal fragments encompassing the ligand-binding domain and the majority of the central modulatory domain together with a C-terminal fragment containing the channel domain and cytosolic tail. Notably, these fragments remain associated with the membrane after initiation of apoptotic cleavage. Furthermore, when recombinant IP3R1 fragments, corresponding to those predicted to be generated by caspase or calpain cleavage, are stably coexpressed in cells, they physically associate and form functional channels. These data provide novel insights regarding the regulation of IP3R1 during proteolysis and provide direct evidence that polypeptide continuity is not required for IP3R activation and Ca(2+) release.
Collapse
Affiliation(s)
- Kamil J Alzayady
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | | | | |
Collapse
|
37
|
Caspase 3 cleavage of the inositol 1,4,5-trisphosphate receptor does not contribute to apoptotic calcium release. Cell Calcium 2012; 53:152-8. [PMID: 23122728 DOI: 10.1016/j.ceca.2012.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 01/26/2023]
Abstract
An important role in the regulation of apoptotic calcium release is played by the ubiquitously expressed family of inositol 1,4,5-trisphosphate receptor (IP(3)R) channels. One model for IP(3)R activation during apoptosis is cleavage by the apoptotic protease caspase 3. Here we show that early elevations in cytosolic calcium during apoptosis do not require caspase 3 activity. We detected a robust increase in calcium levels in response to staurosporine treatment in primary human fibroblasts and HeLa cells in the presence of the caspase inhibitor Z-VAD, indicating that calcium release during the initiation of apoptosis occurs independently of caspase 3. Similar results were obtained with MCF-7 cells which lack caspase 3 expression. Stable expression of caspase 3 in MCF-7 cells and TAT-based transduction of the active recombinant caspase 3 directly into living MCF-7 cells had marginal effects on the early events leading to cytosolic calcium elevations and irreversible commitment to apoptotic cell death. Significantly, blocking IP(3) binding to the IP(3)R with an IP(3) sponge resulted in suppression of staurosporine-induced calcium release and cell death. Collectively, our results suggest that generation of IP(3) is sufficient for the initiation of apoptotic calcium signaling, and caspase 3-mediated truncation of IP(3)R channel is a consequence, not causative, of apoptotic calcium release.
Collapse
|
38
|
Elkoreh G, Blais V, Béliveau E, Guillemette G, Denault JB. Type 1 inositol-1,4,5-trisphosphate receptor is a late substrate of caspases during apoptosis. J Cell Biochem 2012; 113:2775-84. [PMID: 22473799 DOI: 10.1002/jcb.24155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Apoptosis is characterized by the proteolytic cleavage of hundreds of proteins. One of them, the type 1 inositol-1,4,5-trisphosphate receptor (IP(3) R-1), a multimeric receptor located on the endoplasmic reticulum (ER) membrane that is critical to calcium homeostasis, was reported to be cleaved during staurosporine (STS) induced-apoptosis in Jurkat cells. Because the reported cleavage site separates the IP(3) binding site from the channel moiety, its cleavage would shut down a critical signaling pathway that is common to several cellular processes. Here we show that IP(3) R-1 is not cleaved in 293 cells treated with STS, TNFα, Trail, or ultra-violet (UV) irradiation. Further, it is not cleaved in Hela or Jurkat cells induced to undergo apoptosis with Trail, TNFα, or UV. In accordance with previous reports, we demonstrate that it is cleaved in a Jurkat cell line treated with STS. However its cleavage occurs only after poly(ADP-ribose) polymerase (PARP), which cleavage is a hallmark of apoptosis, and p23, a poor caspase-7 substrate, are completely cleaved, suggesting that IP(3) R-1 is a relatively late substrate of caspases. Nevertheless, the receptor is fully accessible to proteolysis in cellulo by ectopically overexpressed caspase-7 or by the tobacco etch virus (TEV) protease. Finally, using recombinant caspase-3 and microsomal fractions enriched in IP(3) R-1, we show that the receptor is a poor caspase-3 substrate. Consequently, we conclude that IP(3) R-1 is not a key death substrate.
Collapse
Affiliation(s)
- Ghadi Elkoreh
- Faculty of Medicine and Health Sciences, Department of Pharmacology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke QC J1H 5N4, Canada
| | | | | | | | | |
Collapse
|
39
|
Abstract
The most well-characterized organelle contact sites are those between the endoplasmic reticulum (ER) and mitochondria. Increased understanding is being gained of how ER-mitochondria contact sites are organized and which factors converge at this interface, some of which may provide a tethering function. The role of the ER-mitochondria junction in coordinating the functions of these two organelles is also becoming clearer, and it has been shown to be involved in the regulation of lipid synthesis, Ca(2+) signalling and the control of mitochondrial biogenesis and intracellular trafficking.
Collapse
|
40
|
Fujimoto T, Shirasawa S. Identification of KRAP-expressing cells and the functional relevance of KRAP to the subcellular localization of IP3R in the stomach and kidney. Int J Mol Med 2012; 30:1287-93. [PMID: 22992961 PMCID: PMC4042864 DOI: 10.3892/ijmm.2012.1126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/20/2012] [Indexed: 12/31/2022] Open
Abstract
KRAS-induced actin-interacting protein (KRAP), originally identified as
one of the deregulated genes expressed in colorectal cancer, participates under
physiological conditions in the regulation of systemic energy homeostasis and of the
exocrine system. We have recently found that KRAP is a molecule associated with inositol
1,4,5-trisphosphate receptor (IP3R) and is critical for the proper subcellular
localization of IP3R in the liver and the pancreas. However, the expression of
KRAP and its precise function in other tissues remain elusive. In this study, we aimed to
identify the KRAP-expressing cells in mouse stomach and kidneys and to examine the
relevance of KRAP expression in the regulation of IP3R localization in these
tissues. In the stomach, double immunohistochemical staining for KRAP and IP3R
demonstrated that KRAP was expressed along with the apical regions in the mucous cells and
the chief cells, and IP3R3 was dominantly co-localized with KRAP in these
cells. Furthermore, IP3R2 was also co-localized with IP3R3 in the
chief cells. It is of note that the proper localization of IP3R3 and
IP3R2 in the chief cells and of IP3R3 in the mucous cells were
significantly abrogated in KRAP-deficient mice. In the kidneys, KRAP was
expressed in both the apical and the basal regions of the proximal tubular cells.
Intriguingly, KRAP deficiency abrogated the localization of
IP3R1 in the proximal tubular cells. Finally, co-immunoprecipitation study in
the stomachs and the kidneys validated the physical association of KRAP with
IP3Rs. These findings demonstrate that KRAP physically associates with
IP3Rs and regulates the proper localization of IP3Rs in the mucous
cells and the chief cells of the stomach and in the proximal tubular cells of the
kidneys.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | | |
Collapse
|
41
|
Matsuzaki H, Fujimoto T, Ota T, Ogawa M, Tsunoda T, Doi K, Hamabashiri M, Tanaka M, Shirasawa S. Tespa1 is a novel inositol 1,4,5-trisphosphate receptor binding protein in T and B lymphocytes. FEBS Open Bio 2012; 2:255-9. [PMID: 23650607 PMCID: PMC3642165 DOI: 10.1016/j.fob.2012.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 07/30/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022] Open
Abstract
Tespa1 has been recently reported to be a critical molecule in T-cell development, however, the precise molecular mechanisms of Tespa1 remain elusive. Here, we demonstrate that Tespa1 shows amino-acid sequence homology to KRAS-induced actin-interacting protein (KRAP), an inositol 1,4,5-trisphosphate receptor (IP3R) binding protein, and that Tespa1 physically associates with IP3R in T and B lymphocytes. Two-consecutive phenylalanine residues (Phe185/Phe186) in Tespa1, which are conserved between Tespa1 and KRAP, are indispensable for the association between Tespa1 and IP3R. These findings suggest that Tespa1 plays critical roles in the immune system through the regulation of the IP3R.
Collapse
Affiliation(s)
- Hiroshi Matsuzaki
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Acute phorbol ester treatment inhibits thapsigargin-induced cell death in porcine aortic smooth muscle cells. Eur J Pharmacol 2012; 686:8-15. [DOI: 10.1016/j.ejphar.2012.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/04/2012] [Accepted: 04/05/2012] [Indexed: 12/11/2022]
|
43
|
Abstract
Ca2+ transfer from endoplasmic reticulum (ER) to mitochondria can trigger apoptotic pathways by inducing release of mitochondrial pro-apoptotic factors. Three different types of inositol 1,4,5-trisphosphate receptor (IP3R) serve to discharge Ca2+ from ER, but possess some peculiarities, especially in apoptosis induction. The anti-apoptotic protein Akt can phosphorylate all IP3R isoforms and protect cells from apoptosis, reducing ER Ca2+ release. However, it has not been elucidated which IP3R subtypes mediate these effects. Here, we show that Akt activation in COS7 cells, which lack of IP3R I, strongly suppresses IP3-mediated Ca2+ release and apoptosis. Conversely, in SH-SY 5Y cells, which are type III-deficient, Akt is unable to modulate ER Ca2+ flux, losing its anti-apoptotic activity. In SH-SY 5Y-expressing subtype III, Akt recovers its protective function on cell death, by reduction of Ca2+ release. Moreover, regulating Ca2+ flux to mitochondria, Akt maintains the mitochondrial integrity and delays the trigger of apoptosis, in a type III-dependent mechanism. These results demonstrate a specific activity of Akt on IP3R III, leading to diminished Ca2+ transfer to mitochondria and protection from apoptosis, suggesting an additional level of cell death regulation mediated by Akt.
Collapse
|
44
|
Fengling M, Qingxiang G, Lijia Z, Wei Z. Influx of extracellular calcium participates in rituximab-enhanced ionizing radiation-induced apoptosis in Raji cells. Toxicol Lett 2012; 209:221-6. [DOI: 10.1016/j.toxlet.2011.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/22/2011] [Accepted: 12/28/2011] [Indexed: 12/31/2022]
|
45
|
Szlufcik K, Missiaen L, Parys JB, Callewaert G, De Smedt H. Uncoupled IP3 receptor can function as a Ca2+-leak channel: cell biological and pathological consequences. Biol Cell 2012; 98:1-14. [PMID: 16354157 DOI: 10.1042/bc20050031] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ca(2+) release via intracellular release channels, IP(3)Rs (inositol 1,4,5-trisphosphate receptors) and RyRs (ryanodine receptors), is perhaps the most ubiquitous and versatile cellular signalling mechanism, and is involved in a vast number of cellular processes. In addition to this classical release pathway there is limited, but yet persistent, information about less well-defined Ca(2+)-leak pathways that may play an important role in the control of the Ca(2+) load of the endo(sarco)plasmic reticulum. The mechanisms responsible for this 'basal' leak are not known, but recent data suggest that both IP(3)Rs and RyRs may also operate as Ca(2+)-leak channels, particularly in pathological conditions. Proteolytic cleavage or biochemical modification (such as hyperphosphorylation or nitrosylation), for example, occurring during conditions of cell stress or apoptosis, can functionally uncouple the cytoplasmic control domains from the channel domain of the receptor. Highly significant information has been obtained from studies of malfunctioning channels in various disorders; for example, RyRs in cardiac malfunction or genetic muscle diseases and IP(3)Rs in neurodegenerative diseases. In this review we aim to summarize the existing information about functionally uncoupled IP(3)R and RyR channels, and to discuss the concept that those channels can participate in Ca(2+)-leak pathways.
Collapse
|
46
|
|
47
|
Abstract
Voltage-dependent anion channels (VDACs) are expressed in three isoforms, with common channeling properties and different roles in cell survival. We show that VDAC1 silencing potentiates apoptotic challenges, whereas VDAC2 has the opposite effect. Although all three VDAC isoforms are equivalent in allowing mitochondrial Ca(2+) loading upon agonist stimulation, VDAC1 silencing selectively impairs the transfer of the low-amplitude apoptotic Ca(2+) signals. Co-immunoprecipitation experiments show that VDAC1, but not VDAC2 and VDAC3, forms complexes with IP(3) receptors, an interaction that is further strengthened by apoptotic stimuli. These data highlight a non-redundant molecular route for transferring Ca(2+) signals to mitochondria in apoptosis.
Collapse
|
48
|
Nguyen YH, Lee KY, Kim TJ, Kim SJ, Kang TM. CD40 Co-stimulation Inhibits Sustained BCR-induced Ca Signaling in Response to Long-term Antigenic Stimulation of Immature B Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:179-87. [PMID: 21860597 DOI: 10.4196/kjpp.2011.15.3.179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 05/30/2011] [Accepted: 05/31/2011] [Indexed: 01/05/2023]
Abstract
Regulation of B cell receptor (BCR)-induced Ca(2+) signaling by CD40 co-stimulation was compared in long-term BCR-stimulated immature (WEHI-231) and mature (Bal-17) B cells. In response to long-term pre-stimulation of immature WEHI-231 cells to α-IgM antibody (0.5~48 hr), the initial transient decrease in BCR-induced [Ca(2+)](i) was followed by spontaneous recovery to control level within 24 hr. The recovery of Ca(2+) signaling in WEHI-231 cells was not due to restoration of internalized receptor but instead to an increase in the levels of PLCγ2 and IP(3)R-3. CD40 co-stimulation of WEHI-231 cells prevented BCR-induced cell cycle arrest and apoptosis, and it strongly inhibited the recovery of BCR-induced Ca(2+) signaling. CD40 co-stimulation also enhanced BCR internalization and reduced expression of PLCγ2 and IP(3)R-3. Pre-treatment of WEHI-231 cells with the antioxidant N-acetyl-L-cysteine (NAC) strongly inhibited CD40-mediated prevention of the recovery of Ca(2+) signaling. In contrast to immature WEHI-231 cells, identical long-term α-IgM pre-stimulation of mature Bal-17 cells abolished the increase in BCR-induced [Ca(2+)](i), regardless of CD40 co-stimulation. These results suggest that CD40-mediated signaling prevents antigen-induced cell cycle arrest and apoptosis of immature B cells through inhibition of sustained BCR-induced Ca(2+) signaling.
Collapse
Affiliation(s)
- Yen Hoang Nguyen
- Department of Physiology, SBRI, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | | | | | | | | |
Collapse
|
49
|
The role of PML in the control of apoptotic cell fate: a new key player at ER-mitochondria sites. Cell Death Differ 2011; 18:1450-6. [PMID: 21475307 DOI: 10.1038/cdd.2011.31] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The development of malignant tumors results from deregulated proliferation or an inability of cells to undergo apoptotic cell death. Experimental works of the past decade have highlighted the importance of calcium (Ca(2+)) in the regulation of apoptosis. Several studies indicate that the Ca(2+) content of the endoplasmic reticulum (ER) determines the cell's sensitivity to apoptotic stress and perturbation of ER Ca(2+) homeostasis appears to be a key component in the development of several pathological situations. Sensitivity to apoptosis depends on the ability of cells to transfer Ca(2+) from the ER to the mitochondria. The physical platform for the interplay between the ER and mitochondria is a domain of the ER called the mitochondria-associated membranes (MAMs). The disruption of these contact sites has profound consequences for cellular function, such as imbalances of intracellular Ca(2+) signaling, cellular stress, and disrupted apoptosis progression. The promyelocytic leukemia (PML) protein has been previously recognized as a critical and essential regulator of multiple apoptotic response. Nevertheless, how PML would exert such broad and fundamental role in apoptosis remained for long time a mystery. In this review, we will discuss how recent results demonstrate that the elusive mechanism whereby the PML tumor suppressor exerts its essential role in apoptosis triggered by Ca(2+)-dependent stimuli can be attributed to its unexpected and fundamental role at MAMs in the control of the functional cross-talk between ER and mitochondria.
Collapse
|
50
|
Shibao K, Fiedler MJ, Nagata J, Minagawa N, Hirata K, Nakayama Y, Iwakiri Y, Nathanson MH, Yamaguchi K. The type III inositol 1,4,5-trisphosphate receptor is associated with aggressiveness of colorectal carcinoma. Cell Calcium 2010; 48:315-23. [PMID: 21075448 PMCID: PMC3572849 DOI: 10.1016/j.ceca.2010.09.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 09/07/2010] [Accepted: 09/08/2010] [Indexed: 01/26/2023]
Abstract
The inositol 1,4,5-trisphosphate receptor (InsP3R) mediates Ca(2+) signaling in epithelia and regulates cellular functions such as secretion, apoptosis and cell proliferation. Loss of one or more InsP3R isoform has been implicated in disease processes such as cholestasis. Here we examined whether gain of expression of InsP3R isoforms also may be associated with development of disease. Expression of all three InsP3R isoforms was evaluated in tissue from colorectal carcinomas surgically resected from 116 patients. Type I and II InsP3Rs were seen in both normal colorectal mucosa and colorectal cancer, while type III InsP3R was observed only in colorectal cancer. Type III InsP3R expression in the advancing margins of tumors correlated with depth of invasion, lymph node metastasis, liver metastasis, and TNM stage. Heavier expression of type III InsP3R also was associated with decreased 5-year survival. shRNA knockdown of type III InsP3R in CACO-2 colon cancer cells enhanced apoptosis, while over-expression of the receptor decreased apoptosis. Thus, type III InsP3R becomes expressed in colon cancer, and its expression level is directly related to aggressiveness of the tumor, which may reflect inhibition of apoptosis by the receptor. These findings suggest a previously unrecognized role for Ca(2+) signaling via this InsP3R isoform in colon cancer.
Collapse
Affiliation(s)
- Kazunori Shibao
- Department of Surgery I, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Michael J. Fiedler
- Digestive Diseases Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jun Nagata
- Department of Surgery I, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Noritaka Minagawa
- Department of Surgery I, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Keiji Hirata
- Department of Nursing, International University of Health and Welfare, Fukuoka, Japan
| | - Yoshifumi Nakayama
- Department of Surgery I, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Yasuko Iwakiri
- Digestive Diseases Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Michael H. Nathanson
- Digestive Diseases Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Koji Yamaguchi
- Department of Surgery I, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| |
Collapse
|