1
|
Zhang X, Tian H, Xie C, Yang Y, Li P, Cheng J. The role and mechanism of vascular wall cell ion channels in vascular fibrosis remodeling. Channels (Austin) 2024; 18:2418128. [PMID: 39425532 PMCID: PMC11492694 DOI: 10.1080/19336950.2024.2418128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/24/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024] Open
Abstract
Fibrosis is usually the final pathological state of many chronic inflammatory diseases and may lead to organ malfunction. Excessive deposition of extracellular matrix (ECM) molecules is a characteristic of most fibrotic tissues. The blood vessel wall contains three layers of membrane structure, including the intima, which is composed of endothelial cells; the media, which is composed of smooth muscle cells; and the adventitia, which is formed by the interaction of connective tissue and fibroblasts. The occurrence and progression of vascular remodeling are closely associated with cardiovascular diseases, and vascular remodeling can alter the original structure and function of the blood vessel. Dysregulation of the composition of the extracellular matrix in blood vessels leads to the continuous advancement of vascular stiffening and fibrosis. Vascular fibrosis reaction leads to excessive deposition of the extracellular matrix in the vascular adventitia, reduces vessel compliance, and ultimately alters key aspects of vascular biomechanics. The pathogenesis of fibrosis in the vasculature and strategies for its reversal have become interesting and important challenges. Ion channels are widely expressed in the cardiovascular system; they regulate blood pressure, maintain cardiovascular function homeostasis, and play important roles in ion transport, cell differentiation, proliferation. In blood vessels, different types of ion channels in fibroblasts, smooth muscle cells and endothelial cells may be relevant mediators of the development of fibrosis in organs or tissues. This review discusses the known roles of ion channels in vascular fibrosis remodeling and discusses potential therapeutic targets for regulating remodeling and repair after vascular injury.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Hai Tian
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Cheng Xie
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Public Center of Experimental Technology, Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Djordjevic S, Itzykson R, Hague F, Lebon D, Legrand J, Ouled‐Haddou H, Jedraszak G, Harbonnier J, Collet L, Paubelle E, Marolleau J, Garçon L, Boyer T. STIM2 is involved in the regulation of apoptosis and the cell cycle in normal and malignant monocytic cells. Mol Oncol 2024; 18:1571-1592. [PMID: 38234211 PMCID: PMC11161727 DOI: 10.1002/1878-0261.13584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/28/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Calcium is a ubiquitous messenger that regulates a wide range of cellular functions, but its involvement in the pathophysiology of acute myeloid leukemia (AML) is not widely investigated. Here, we identified, from an analysis of The Cancer Genome Atlas and genotype-tissue expression databases, stromal interaction molecule 2 (STIM2) as being highly expressed in AML with monocytic differentiation and negatively correlated with overall survival. This was confirmed on a validation cohort of 407 AML patients. We then investigated the role of STIM2 in cell proliferation, differentiation, and survival in two leukemic cell lines with monocytic potential and in normal hematopoietic stem cells. STIM2 expression increased at the RNA and protein levels upon monocyte differentiation. Phenotypically, STIM2 knockdown drastically inhibited cell proliferation and induced genomic stress with DNA double-strand breaks, as shown by increased levels of phosphorylate histone H2AXγ (p-H2AXγ), followed by activation of the cellular tumor antigen p53 pathway, decreased expression of cell cycle regulators such as cyclin-dependent kinase 1 (CDK1)-cyclin B1 and M-phase inducer phosphatase 3 (CDC25c), and a decreased apoptosis threshold with a low antiapoptotic/proapoptotic protein ratio. Our study reports STIM2 as a new actor regulating genomic stability and p53 response in terms of cell cycle and apoptosis of human normal and malignant monocytic cells.
Collapse
Affiliation(s)
| | - Raphaël Itzykson
- Département Hématologie et ImmunologieHôpital Saint‐Louis, Assistance Publique‐Hôpitaux de ParisFrance
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRSUniversité Paris CitéFrance
| | - Frédéric Hague
- Laboratoire de Physiologie Cellulaire et Moléculaire UR4667Université Picardie Jules VerneAmiensFrance
| | - Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Julien Legrand
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Laboratoire de Génétique ConstitutionnelleCHU Amiens‐PicardieFrance
| | | | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Etienne Paubelle
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Clinique et de Thérapie CellulaireCHU Amiens‐PicardieFrance
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie BiologiqueCHU Amiens‐PicardieFrance
| |
Collapse
|
3
|
Scaviner J, Bagacean C, Christian B, Renaudineau Y, Mignen O, Abdoul-Azize S. Blocking Orai1 constitutive activity inhibits B-cell cancer migration and synergistically acts with drugs to reduce B-CLL cell survival. Eur J Pharmacol 2024; 971:176515. [PMID: 38547958 DOI: 10.1016/j.ejphar.2024.176515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/10/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024]
Abstract
Orai1 channel capacity to control store-operated Ca2+ entry (SOCE) and B-cell functions is poorly understood and more specifically in B-cell cancers, including human lymphoma and leukemia. As compared to normal B-cells, Orai1 is overexpressed in B-chronic lymphocytic leukemia (B-CLL) and contributes in resting B-CLL to mediate an elevated basal Ca2+ level through a constitutive Ca2+ entry, and in BCR-activated B-cell to regulate the Ca2+ signaling response. Such observations were confirmed in human B-cell lymphoma and leukemia lines, including RAMOS, JOK-1, MEC-1 and JVM-3 cells. Next, the use of pharmacological Orai1 inhibitors (GSK-7975 A and Synta66) blocks constitutive Ca2+ entry and in turn affects B-cell cancer (primary and cell lines) survival and migration, controls cell cycle, and induces apoptosis through a mitochondrial and caspase-3 independent pathway. Finally, the added value of Orai1 inhibitors in combination with B-CLL drugs (ibrutinib, idelalisib, rituximab, and venetoclax) on B-CLL survival was tested, showing an additive/synergistic effect including in the B-cell cancer lines. To conclude, this study highlights the pathophysiological role of the Ca2+ channel Orai1 in B-cell cancers, and pave the way for the use of ORAI1 modulators as a plausible therapeutic strategy.
Collapse
Affiliation(s)
- Julien Scaviner
- INSERM UMR1227, Université de Bretagne Occidentale, F-29200 Brest, France
| | - Cristina Bagacean
- INSERM UMR1227, Université de Bretagne Occidentale, F-29200 Brest, France
| | - Berthou Christian
- INSERM UMR1227, Université de Bretagne Occidentale, F-29200 Brest, France
| | - Yves Renaudineau
- INSERM UMR1227, Université de Bretagne Occidentale, F-29200 Brest, France
| | - Olivier Mignen
- INSERM UMR1227, Université de Bretagne Occidentale, F-29200 Brest, France
| | | |
Collapse
|
4
|
Macias‐Diaz A, Lopez JJ, Bravo M, Jardín I, Garcia‐Jimenez WL, Blanco‐Blanco FJ, Cerrato R, Rosado JA. Postbiotics of Lacticaseibacillus paracasei CECT 9610 and Lactiplantibacillus plantarum CECT 9608 attenuates store-operated calcium entry and FAK phosphorylation in colorectal cancer cells. Mol Oncol 2024; 18:1123-1142. [PMID: 38514909 PMCID: PMC11076996 DOI: 10.1002/1878-0261.13629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024] Open
Abstract
Store-operated Ca2+ entry (SOCE) is a major mechanism for Ca2+ influx in colorectal cancer (CRC) cells. This mechanism, regulated by the filling state of the intracellular Ca2+ stores, is mediated by the endoplasmic reticulum Ca2+ sensors of the stromal interaction molecules (STIM) family [stromal interaction molecule 1 (STIM1) and STIM2] and the Ca2+-release-activated Ca2+ channels constituted by Orai family members, with predominance of calcium release-activated calcium channel protein 1 (Orai1). CRC cells exhibit enhanced SOCE due to remodeling of the expression of the key SOCE molecular components. The enhanced SOCE supports a variety of cancer hallmarks. Here, we show that treatment of the colorectal adenocarcinoma cell lines HT-29 and Caco-2 with inanimate Lacticaseibacillus paracasei (CECT9610) and Lactiplantibacillus plantarum (CECT9608) attenuates SOCE, although no detectable effect is seen on SOCE in normal colon mucosa cells. The effect of Lacticaseibacillus paracasei and Lactiplantibacillus plantarum postbiotics was mediated by downregulation of Orai1 and STIM1, while the expression levels of Orai3 and STIM2 remained unaltered. Treatment of HT-29 and Caco-2 cells with inanimate Lacticaseibacillus paracasei and Lactiplantibacillus plantarum impairs in vitro migration by a mechanism likely involving attenuation of focal adhesion kinase (FAK) tyrosine phosphorylation. Cell treatment with the Orai1 inhibitor synta-66 attenuates SOCE and prevents any further effect of Lacticaseibacillus paracasei and Lactiplantibacillus plantarum postbiotics. Together, our results indicate for the first time that Lacticaseibacillus paracasei and Lactiplantibacillus plantarum postbiotics selectively exert negative effects on Ca2+ influx through SOCE in colorectal adenocarcinoma cell lines, providing evidence for an attractive strategy against CRC.
Collapse
Affiliation(s)
- Alvaro Macias‐Diaz
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB)Universidad de ExtremaduraCáceresSpain
| | - Jose J. Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB)Universidad de ExtremaduraCáceresSpain
| | - Maria Bravo
- Innovación en Gestión y Conservación de Ungulados S.LCáceresSpain
| | - Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB)Universidad de ExtremaduraCáceresSpain
| | | | | | - Rosario Cerrato
- Innovación en Gestión y Conservación de Ungulados S.LCáceresSpain
| | - Juan A. Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB)Universidad de ExtremaduraCáceresSpain
| |
Collapse
|
5
|
Lebon D, Collet L, Djordjevic S, Gomila C, Ouled‐Haddou H, Platon J, Demont Y, Marolleau J, Caulier A, Garçon L. PIEZO1 is essential for the survival and proliferation of acute myeloid leukemia cells. Cancer Med 2024; 13:e6984. [PMID: 38334477 PMCID: PMC10854442 DOI: 10.1002/cam4.6984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/06/2024] [Accepted: 01/19/2024] [Indexed: 02/10/2024] Open
Abstract
INTRODUCTION Leukemogenesis is a complex process that interconnects tumoral cells with their microenvironment, but the effect of mechanosensing in acute myeloid leukemia (AML) blasts is poorly known. PIEZO1 perceives and transmits the constraints of the environment to human cells by acting as a non-selective calcium channel, but very little is known about its role in leukemogenesis. RESULTS For the first time, we show that PIEZO1 is preferentially expressed in healthy hematopoietic stem and progenitor cells in human hematopoiesis, and globally overexpressed in AML cells. In AML subtypes, PIEZO1 expression associates with favorable outcomes as better overall (OS) and disease-free survival (DFS). If PIEZO1 is expressed and functional in THP1 leukemic myeloid cell line, its chemical activation doesn't impact the proliferation, differentiation, nor survival of cells. However, the downregulation of PIEZO1 expression dramatically reduces the proliferation and the survival of THP1 cells. We show that PIEZO1 knock-down blocks the cell cycle in G0/G1 phases of AML cells, impairs the DNA damage response pathways, and critically increases cell death by triggering extrinsic apoptosis pathways. CONCLUSIONS Altogether, our results reveal a new role for PIEZO1 mechanosensing in the survival and proliferation of leukemic blasts, which could pave the way for new therapeutic strategies to target AML cells.
Collapse
Affiliation(s)
- Delphine Lebon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Hématologie Clinique et Thérapie Cellulaire, CHU Amiens‐PicardieAmiensFrance
| | - Louison Collet
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Hématologie Clinique et Thérapie Cellulaire, CHU Amiens‐PicardieAmiensFrance
| | | | - Cathy Gomila
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | - Jessica Platon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Yohann Demont
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Biologie, CHU Amiens‐PicardieAmiensFrance
| | - Jean‐Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Hématologie Clinique et Thérapie Cellulaire, CHU Amiens‐PicardieAmiensFrance
| | - Alexis Caulier
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Division of Hematology/Oncology Boston Children's HospitalBostonMassachusettsUSA
- Department of Medical and Population GeneticsThe Broad Institute of Harvard and MITCambridgeMassachusettsUSA
| | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
- Service d'Hématologie Biologie, CHU Amiens‐PicardieAmiensFrance
| |
Collapse
|
6
|
Abdelnaby AE, Trebak M. Store-Operated Ca 2+ Entry in Fibrosis and Tissue Remodeling. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241291374. [PMID: 39659877 PMCID: PMC11629433 DOI: 10.1177/25152564241291374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/29/2024] [Accepted: 09/27/2024] [Indexed: 12/12/2024]
Abstract
Fibrosis is a pathological condition characterized by excessive tissue deposition of extracellular matrix (ECM) components, leading to scarring and impaired function across multiple organ systems. This complex process is mediated by a dynamic interplay between cell types, including myofibroblasts, fibroblasts, immune cells, epithelial cells, and endothelial cells, each contributing distinctively through various signaling pathways. Critical to the regulatory mechanisms involved in fibrosis is store-operated calcium entry (SOCE), a calcium entry pathway into the cytosol active at the endoplasmic reticulum-plasma membrane contact sites and common to all cells. This review addresses the multifactorial nature of fibrosis with a focus on the pivotal roles of different cell types. We highlight the essential functions of myofibroblasts in ECM production, the transformation of fibroblasts, and the participation of immune cells in modulating the fibrotic landscape. We emphasize the contributions of SOCE in these different cell types to fibrosis, by exploring the involvement of SOCE in cellular functions such as proliferation, migration, secretion, and inflammatory responses. The examination of the cellular and molecular mechanisms of fibrosis and the role of SOCE in these mechanisms offers the potential of targeting SOCE as a therapeutic strategy for mitigating or reversing fibrosis.
Collapse
Affiliation(s)
- Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
8
|
Immanuel T, Li J, Green TN, Bogdanova A, Kalev-Zylinska ML. Deregulated calcium signaling in blood cancer: Underlying mechanisms and therapeutic potential. Front Oncol 2022; 12:1010506. [PMID: 36330491 PMCID: PMC9623116 DOI: 10.3389/fonc.2022.1010506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Intracellular calcium signaling regulates diverse physiological and pathological processes. In solid tumors, changes to calcium channels and effectors via mutations or changes in expression affect all cancer hallmarks. Such changes often disrupt transport of calcium ions (Ca2+) in the endoplasmic reticulum (ER) or mitochondria, impacting apoptosis. Evidence rapidly accumulates that this is similar in blood cancer. Principles of intracellular Ca2+ signaling are outlined in the introduction. We describe different Ca2+-toolkit components and summarize the unique relationship between extracellular Ca2+ in the endosteal niche and hematopoietic stem cells. The foundational data on Ca2+ homeostasis in red blood cells is discussed, with the demonstration of changes in red blood cell disorders. This leads to the role of Ca2+ in neoplastic erythropoiesis. Then we expand onto the neoplastic impact of deregulated plasma membrane Ca2+ channels, ER Ca2+ channels, Ca2+ pumps and exchangers, as well as Ca2+ sensor and effector proteins across all types of hematologic neoplasms. This includes an overview of genetic variants in the Ca2+-toolkit encoding genes in lymphoid and myeloid cancers as recorded in publically available cancer databases. The data we compiled demonstrate that multiple Ca2+ homeostatic mechanisms and Ca2+ responsive pathways are altered in hematologic cancers. Some of these alterations may have genetic basis but this requires further investigation. Most changes in the Ca2+-toolkit do not appear to define/associate with specific disease entities but may influence disease grade, prognosis, treatment response, and certain complications. Further elucidation of the underlying mechanisms may lead to novel treatments, with the aim to tailor drugs to different patterns of deregulation. To our knowledge this is the first review of its type in the published literature. We hope that the evidence we compiled increases awareness of the calcium signaling deregulation in hematologic neoplasms and triggers more clinical studies to help advance this field.
Collapse
Affiliation(s)
- Tracey Immanuel
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jixia Li
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan City, China
| | - Taryn N. Green
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Haematology Laboratory, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
9
|
Lewuillon C, Guillemette A, Titah S, Shaik FA, Jouy N, Labiad O, Farfariello V, Laguillaumie MO, Idziorek T, Barthélémy A, Peyrouze P, Berthon C, Tarhan MC, Cheok M, Quesnel B, Lemonnier L, Touil Y. Involvement of ORAI1/SOCE in Human AML Cell Lines and Primary Cells According to ABCB1 Activity, LSC Compartment and Potential Resistance to Ara-C Exposure. Int J Mol Sci 2022; 23:ijms23105555. [PMID: 35628366 PMCID: PMC9141756 DOI: 10.3390/ijms23105555] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy with a high risk of relapse. This issue is associated with the development of mechanisms leading to drug resistance that are not yet fully understood. In this context, we previously showed the clinical significance of the ATP binding cassette subfamily B-member 1 (ABCB1) in AML patients, namely its association with stemness markers and an overall worth prognosis. Calcium signaling dysregulations affect numerous cellular functions and are associated with the development of the hallmarks of cancer. However, in AML, calcium-dependent signaling pathways remain poorly investigated. With this study, we show the involvement of the ORAI1 calcium channel in store-operated calcium entry (SOCE), the main calcium entry pathway in non-excitable cells, in two representative human AML cell lines (KG1 and U937) and in primary cells isolated from patients. Moreover, our data suggest that in these models, SOCE varies according to the differentiation status, ABCB1 activity level and leukemic stem cell (LSC) proportion. Finally, we present evidence that ORAI1 expression and SOCE amplitude are modulated during the establishment of an apoptosis resistance phenotype elicited by the chemotherapeutic drug Ara-C. Our results therefore suggest ORAI1/SOCE as potential markers of AML progression and drug resistance apparition.
Collapse
Affiliation(s)
- Clara Lewuillon
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Aurélie Guillemette
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Sofia Titah
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Faruk Azam Shaik
- Institut de Recherche sur le Cancer de Lille (IRCL), F-59000 Lille, France;
- LIMMS/CNRS-IIS IRL2820, The University of Tokyo, Tokyo 153-8505, Japan;
| | - Nathalie Jouy
- UMS 2014/US41 Plateformes Lilloises En Biologie Et Sante, Université de Lille, F-59000 Lille, France;
| | - Ossama Labiad
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Valerio Farfariello
- Inserm, U1003-PHYCEL-Physiologie Cellulaire, Université de Lille, F-59000 Lille, France;
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, F-59655 Villeneuve d’Ascq, France
| | - Marie-Océane Laguillaumie
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Thierry Idziorek
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Adeline Barthélémy
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Pauline Peyrouze
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Céline Berthon
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Mehmet Cagatay Tarhan
- LIMMS/CNRS-IIS IRL2820, The University of Tokyo, Tokyo 153-8505, Japan;
- CNRS, Centrale Lille, Junia, Université Polytechnique Hauts-de-France, UMR 8520—IEMN—Institut d’Electronique de Microélectronique et de Nanotechnologie, Université de Lille, F-59000 Lille, France
| | - Meyling Cheok
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Bruno Quesnel
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
| | - Loïc Lemonnier
- Inserm, U1003-PHYCEL-Physiologie Cellulaire, Université de Lille, F-59000 Lille, France;
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, F-59655 Villeneuve d’Ascq, France
- Correspondence: loï (L.L.); (Y.T.)
| | - Yasmine Touil
- CNRS, Inserm, CHU Lille, UMR 9020, UMR-S 1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, Université de Lille, F-59000 Lille, France; (C.L.); (A.G.); (S.T.); (O.L.); (M.-O.L.); (T.I.); (A.B.); (P.P.); (C.B.); (M.C.); (B.Q.)
- Correspondence: loï (L.L.); (Y.T.)
| |
Collapse
|
10
|
Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells. Cells 2022; 11:cells11081332. [PMID: 35456011 PMCID: PMC9032688 DOI: 10.3390/cells11081332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 12/25/2022] Open
Abstract
Tumors are composed by a heterogeneous population of cells. Among them, a sub-population of cells, termed cancer stem cells, exhibit stemness features, such as self-renewal capabilities, disposition to differentiate to a more proliferative state, and chemotherapy resistance, processes that are all mediated by Ca2+. Ca2+ homeostasis is vital for several physiological processes, and alterations in the patterns of expressions of the proteins and molecules that modulate it have recently become a cancer hallmark. Store-operated Ca2+ entry is a major mechanism for Ca2+ entry from the extracellular medium in non-excitable cells that leads to increases in the cytosolic Ca2+ concentration required for several processes, including cancer stem cell properties. Here, we focus on the participation of STIM, Orai, and TRPC proteins, the store-operated Ca2+ entry key components, in cancer stem cell biology and tumorigenesis.
Collapse
|
11
|
Lewuillon C, Laguillaumie MO, Quesnel B, Idziorek T, Touil Y, Lemonnier L. Put in a “Ca2+ll” to Acute Myeloid Leukemia. Cells 2022; 11:cells11030543. [PMID: 35159351 PMCID: PMC8834247 DOI: 10.3390/cells11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by genetic aberrations in myeloid primitive cells (blasts) which lead to their defective maturation/function and their proliferation in the bone marrow (BM) and blood of affected individuals. Current intensive chemotherapy protocols result in complete remission in 50% to 80% of AML patients depending on their age and the AML type involved. While alterations in calcium signaling have been extensively studied in solid tumors, little is known about the role of calcium in most hematologic malignancies, including AML. Our purpose with this review is to raise awareness about this issue and to present (i) the role of calcium signaling in AML cell proliferation and differentiation and in the quiescence of hematopoietic stem cells; (ii) the interplay between mitochondria, metabolism, and oxidative stress; (iii) the effect of the BM microenvironment on AML cell fate; and finally (iv) the mechanism by which chemotherapeutic treatments modify calcium homeostasis in AML cells.
Collapse
Affiliation(s)
- Clara Lewuillon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Marie-Océane Laguillaumie
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Thierry Idziorek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Yasmine Touil
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (C.L.); (M.-O.L.); (B.Q.); (T.I.); (Y.T.)
| | - Loïc Lemonnier
- Univ. Lille, Inserm, U1003—PHYCEL—Physiologie Cellulaire, F-59000 Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, F-59655 Villeneuve d’Ascq, France
- Correspondence:
| |
Collapse
|
12
|
Orai2 Modulates Store-Operated Ca 2+ Entry and Cell Cycle Progression in Breast Cancer Cells. Cancers (Basel) 2021; 14:cancers14010114. [PMID: 35008277 PMCID: PMC8749845 DOI: 10.3390/cancers14010114] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/07/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is a heterogeneous disease from the histological and molecular expression point of view, and this heterogeneity determines cancer aggressiveness. Store-operated Ca2+ entry (SOCE), a major mechanism for Ca2+ entry in non-excitable cells, is significantly remodeled in cancer cells and plays an important role in the development and support of different cancer hallmarks. The store-operated CRAC (Ca2+ release-activated Ca2+) channels are predominantly comprised of Orai1 but the participation of Orai2 and Orai3 subunits has been reported to modulate the magnitude of Ca2+ responses. Here we provide evidence for a heterogeneous expression of Orai2 among different breast cancer cell lines. In the HER2 and triple negative breast cancer cell lines SKBR3 and BT20, respectively, where the expression of Orai2 was greater, Orai2 modulates the magnitude of SOCE and sustain Ca2+ oscillations in response to carbachol. Interestingly, in these cells Orai2 modulates the activation of NFAT1 and NFAT4 in response to high and low agonist concentrations. Finally, we have found that, in cells with high Orai2 expression, Orai2 knockdown leads to cell cycle arrest at the G0-G1 phase and decreases apoptosis resistance upon cisplatin treatment. Altogether, these findings indicate that, in breast cancer cells with a high Orai2 expression, Orai2 plays a relevant functional role in agonist-evoked Ca2+ signals, cell proliferation and apoptosis resistance.
Collapse
|
13
|
Bokhobza A, Ziental-Gelus N, Allart L, Iamshanova O, Vanden Abeele F. Impact of SOCE Abolition by ORAI1 Knockout on the Proliferation, Adhesion, and Migration of HEK-293 Cells. Cells 2021; 10:3016. [PMID: 34831241 PMCID: PMC8616168 DOI: 10.3390/cells10113016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
Store-operated calcium entry (SOCE) provided through channels formed by ORAI proteins is a major regulator of several cellular processes. In immune cells, it controls fundamental processes such as proliferation, cell adhesion, and migration, while in cancer, SOCE and ORAI1 gene expression are dysregulated and lead to abnormal migration and/or cell proliferation. In the present study, we used the CRISPR/Cas9 technique to delete the ORAI1 gene and to identify its role in proliferative and migrative properties of the model cell line HEK-293. We showed that ORAI1 deletion greatly reduced SOCE. Thereby, we found that this decrease and the absence of ORAI1 protein did not affect HEK-293 proliferation. In addition, we determined that ORAI1 suppression did not affect adhesive properties but had a limited impact on HEK-293 migration. Overall, we showed that ORAI1 and SOCE are largely dispensable for cellular proliferation, migration, and cellular adhesion of HEK-293 cells. Thus, despite its importance in providing Ca2+ entry in non-excitable cells, our results indicate that the lack of SOCE does not deeply impact HEK-293 cells. This finding suggests the existence of compensatory mechanism enabling the maintenance of their physiological function.
Collapse
Affiliation(s)
- Alexandre Bokhobza
- Inserm U1003, Laboratory of Cell Physiology, Université de Lille, 59650 Villeneuve d’Ascq, France; (N.Z.-G.); (L.A.); (O.I.)
| | | | | | | | - Fabien Vanden Abeele
- Inserm U1003, Laboratory of Cell Physiology, Université de Lille, 59650 Villeneuve d’Ascq, France; (N.Z.-G.); (L.A.); (O.I.)
| |
Collapse
|
14
|
Xu F, Wang Y, Gao H, Zhang X, Hu Y, Han T, Shen B, Zhang L, Wu Q. X-Ray Causes mRNA Transcripts Change to Enhance Orai2-Mediated Ca 2+ Influx in Rat Brain Microvascular Endothelial Cells. Front Mol Biosci 2021; 8:646730. [PMID: 34595206 PMCID: PMC8477418 DOI: 10.3389/fmolb.2021.646730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/05/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Radiation-induced brain injury is a serious and treatment-limiting complication of brain radiation therapy. Although endothelial cell dysfunction plays a critical role in the development of this pathogenesis, the underlying molecular mechanisms remain elusive. Methods: Primary cultured rat brain microvascular endothelial cells (BMECs) were divided into five groups without or with exposure of x-rays delivered at 5 Gy or 20 Gy. For the irradiated groups, cells were continued to cultivate for 12 or 24 h after being irradiated. Then the mRNA libraries of each group were established and applied for next-generation sequencing. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were conducted to analyze the sequencing results. Quantitative polymerase chain reaction, western blotting, cck8 assay and intracellular calcium concentration assays were conducted to analyze the role of Orai2-associated SOCE in x-ray induced cellular injury. Results: In total, 3,005 transcripts in all the four x-ray-exposed groups of BMECs showed expression level changes compared with controls. With the dose of x-ray augment and the following cultured time extension, the numbers of differentially expressed genes (DEGs) increased significantly in BMECs. Venn diagrams identified 40 DEGs common to all four exposure groups. Functional pathway enrichment analyses indicated that those 40 DEGs were enriched in the calcium signaling pathway. Among those 40 DEGs, mRNA and protein expression levels of Orai2 were significantly upregulated for 24 h. Similarly, calcium influx via store-operated calcium entry, which is modulated by Orai2, was also significantly increased for 24 h in x-ray-exposed BMECs. Moreover, the change in SOCE was suppressed by btp-2, which is a non-selective inhibitor of Orai. Additionally, x-ray exposure induced a significant decrease of proliferation in BMECs in the dose- and time-dependent manner. Conclusion: These findings provide evidence for molecular mechanisms underlying BMECs dysfunction in development of radiation-induced brain injury and suggest new approaches for therapeutic targets.
Collapse
Affiliation(s)
- Fangfang Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Wang
- Department of Otolaryngology-Head and Neck Surgery, Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China.,School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Huiwen Gao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinchen Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Hu
- Department of Otolaryngology-Head and Neck Surgery, Lu'an People's Hospital, Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, China
| | - Tingting Han
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lesha Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qibing Wu
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Sharma A, Ramena GT, Elble RC. Advances in Intracellular Calcium Signaling Reveal Untapped Targets for Cancer Therapy. Biomedicines 2021; 9:1077. [PMID: 34572262 PMCID: PMC8466575 DOI: 10.3390/biomedicines9091077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Ca2+ distribution is a tightly regulated process. Numerous Ca2+ chelating, storage, and transport mechanisms are required to maintain normal cellular physiology. Ca2+-binding proteins, mainly calmodulin and calbindins, sequester free intracellular Ca2+ ions and apportion or transport them to signaling hubs needing the cations. Ca2+ channels, ATP-driven pumps, and exchangers assist the binding proteins in transferring the ions to and from appropriate cellular compartments. Some, such as the endoplasmic reticulum, mitochondria, and lysosomes, act as Ca2+ repositories. Cellular Ca2+ homeostasis is inefficient without the active contribution of these organelles. Moreover, certain key cellular processes also rely on inter-organellar Ca2+ signaling. This review attempts to encapsulate the structure, function, and regulation of major intracellular Ca2+ buffers, sensors, channels, and signaling molecules before highlighting how cancer cells manipulate them to survive and thrive. The spotlight is then shifted to the slow pace of translating such research findings into anticancer therapeutics. We use the PubMed database to highlight current clinical studies that target intracellular Ca2+ signaling. Drug repurposing and improving the delivery of small molecule therapeutics are further discussed as promising strategies for speeding therapeutic development in this area.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Grace T. Ramena
- Department of Aquaculture, University of Arkansas, Pine Bluff, AR 71601, USA;
| | - Randolph C. Elble
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| |
Collapse
|
16
|
Cross-Talk Between the Adenylyl Cyclase/cAMP Pathway and Ca 2+ Homeostasis. Rev Physiol Biochem Pharmacol 2021; 179:73-116. [PMID: 33398503 DOI: 10.1007/112_2020_55] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cyclic AMP and Ca2+ are the first second or intracellular messengers identified, unveiling the cellular mechanisms activated by a plethora of extracellular signals, including hormones. Cyclic AMP generation is catalyzed by adenylyl cyclases (ACs), which convert ATP into cAMP and pyrophosphate. By the way, Ca2+, as energy, can neither be created nor be destroyed; Ca2+ can only be transported, from one compartment to another, or chelated by a variety of Ca2+-binding molecules. The fine regulation of cytosolic concentrations of cAMP and free Ca2+ is crucial in cell function and there is an intimate cross-talk between both messengers to fine-tune the cellular responses. Cancer is a multifactorial disease resulting from a combination of genetic and environmental factors. Frequent cases of cAMP and/or Ca2+ homeostasis remodeling have been described in cancer cells. In those tumoral cells, cAMP and Ca2+ signaling plays a crucial role in the development of hallmarks of cancer, including enhanced proliferation and migration, invasion, apoptosis resistance, or angiogenesis. This review summarizes the cross-talk between the ACs/cAMP and Ca2+ intracellular pathways with special attention to the functional and reciprocal regulation between Orai1 and AC8 in normal and cancer cells.
Collapse
|
17
|
Sánchez-Collado J, López JJ, Rosado JA. The Orai1-AC8 Interplay: How Breast Cancer Cells Escape from Orai1 Channel Inactivation. Cells 2021; 10:1308. [PMID: 34070268 PMCID: PMC8225208 DOI: 10.3390/cells10061308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
The interplay between the Ca2+-sensitive adenylyl cyclase 8 (AC8) and Orai1 channels plays an important role both in the activation of the cAMP/PKA signaling and the modulation of Orai1-dependent Ca2+ signals. AC8 interacts with a N-terminal region that is exclusive to the Orai1 long variant, Orai1α. The interaction between both proteins allows the Ca2+ that enters the cell through Orai1α to activate the generation of cAMP by AC8. Subsequent PKA activation results in Orai1α inactivation by phosphorylation at serine-34, thus shaping Orai1-mediated cellular functions. In breast cancer cells, AC8 plays a relevant role supporting a variety of cancer hallmarks, including proliferation and migration. Breast cancer cells overexpress AC8, which shifts the AC8-Orai1 stoichiometry in favor of the former and leads to the impairment of PKA-dependent Orai1α inactivation. This mechanism contributes to the enhanced SOCE observed in triple-negative breast cancer cells. This review summarizes the functional interaction between AC8 and Orai1α in normal and breast cancer cells and its relevance for different cancer features.
Collapse
Affiliation(s)
| | - José J. López
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| | - Juan A. Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain;
| |
Collapse
|
18
|
Maggi F, Morelli MB, Nabissi M, Marinelli O, Zeppa L, Aguzzi C, Santoni G, Amantini C. Transient Receptor Potential (TRP) Channels in Haematological Malignancies: An Update. Biomolecules 2021; 11:biom11050765. [PMID: 34065398 PMCID: PMC8160608 DOI: 10.3390/biom11050765] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are improving their importance in different cancers, becoming suitable as promising candidates for precision medicine. Their important contribution in calcium trafficking inside and outside cells is coming to light from many papers published so far. Encouraging results on the correlation between TRP and overall survival (OS) and progression-free survival (PFS) in cancer patients are available, and there are as many promising data from in vitro studies. For what concerns haematological malignancy, the role of TRPs is still not elucidated, and data regarding TRP channel expression have demonstrated great variability throughout blood cancer so far. Thus, the aim of this review is to highlight the most recent findings on TRP channels in leukaemia and lymphoma, demonstrating their important contribution in the perspective of personalised therapies.
Collapse
Affiliation(s)
- Federica Maggi
- Department of Molecular Medicine, Sapienza University, 00185 Rome, Italy;
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Maria Beatrice Morelli
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Massimo Nabissi
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Oliviero Marinelli
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Laura Zeppa
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Cristina Aguzzi
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Giorgio Santoni
- Immunopathology Laboratory, School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (M.B.M.); (M.N.); (O.M.); (L.Z.); (C.A.); (G.S.)
| | - Consuelo Amantini
- Immunopathology Laboratory, School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
- Correspondence: ; Tel.: +30-0737403312
| |
Collapse
|
19
|
Shapovalov G, Gordienko D, Prevarskaya N. Store operated calcium channels in cancer progression. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:123-168. [PMID: 34392928 DOI: 10.1016/bs.ircmb.2021.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent decades cancer emerged as one of the leading causes of death in the developed countries, with some types of cancer contributing to the top 10 causes of death on the list of the World Health Organization. Carcinogenesis, a malignant transformation causing formation of tumors in normal tissues, is associated with changes in the cell cycle caused by suppression of signaling pathways leading to cell death and facilitation of those enhancing proliferation. Further progression of cancer, during which benign tumors acquire more aggressive phenotypes, is characterized by metastatic dissemination through the body driven by augmented motility and invasiveness of cancer cells. All these processes are associated with alterations in calcium homeostasis in cancer cells, which promote their proliferation, motility and invasion, and dissuade cell death or cell cycle arrest. Remodeling of store-operated calcium entry (SOCE), one of the major pathways regulating intracellular Ca2+ concentration ([Ca2+]i), manifests a key event in many of these processes. This review systematizes current knowledge on the mechanisms recruiting SOCE-related proteins in carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- George Shapovalov
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologiesa, University of Lille, Villeneuve d'Ascq, France.
| | - Dmitri Gordienko
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologiesa, University of Lille, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologiesa, University of Lille, Villeneuve d'Ascq, France
| |
Collapse
|
20
|
Cheng Z, Dai Y, Huang W, Zhong Q, Zhu P, Zhang W, Wu Z, Lin Q, Zhu H, Cui L, Qian T, Deng C, Fu L, Liu Y, Zeng T. Prognostic Value of MicroRNA-20b in Acute Myeloid Leukemia. Front Oncol 2021; 10:553344. [PMID: 33680910 PMCID: PMC7930740 DOI: 10.3389/fonc.2020.553344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous disease that requires fine-grained risk stratification for the best prognosis of patients. As a class of small non-coding RNAs with important biological functions, microRNAs play a crucial role in the pathogenesis of AML. To assess the prognostic impact of miR-20b on AML in the presence of other clinical and molecular factors, we screened 90 AML patients receiving chemotherapy only and 74 also undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) from the Cancer Genome Atlas (TCGA) database. In the chemotherapy-only group, high miR-20b expression subgroup had shorter event-free survival (EFS) and overall survival (OS, both P < 0.001); whereas, there were no significant differences in EFS and OS between high and low expression subgroups in the allo-HSCT group. Then we divided all patients into high and low expression groups based on median miR-20b expression level. In the high expression group, patients treated with allo-HSCT had longer EFS and OS than those with chemotherapy alone (both P < 0.01); however, there were no significant differences in EFS and OS between different treatment subgroups in the low expression group. Further analysis showed that miR-20b was negatively correlated with genes in “ribosome,” “myeloid leukocyte mediated immunity,” and “DNA replication” signaling pathways. ORAI2, the gene with the strongest correlation with miR-20b, also had significant prognostic value in patients undergoing chemotherapy but not in the allo-HSCT group. In conclusion, our findings suggest that high miR-20b expression is a poor prognostic indicator for AML, but allo-HSCT may override its prognostic impact.
Collapse
Affiliation(s)
- Zhiheng Cheng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Dai
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingfu Zhong
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pei Zhu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjuan Zhang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhihua Wu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qing Lin
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huoyan Zhu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cong Deng
- Department of Clinical laboratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tiansheng Zeng
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
21
|
Alharbi A, Zhang Y, Parrington J. Deciphering the Role of Ca 2+ Signalling in Cancer Metastasis: From the Bench to the Bedside. Cancers (Basel) 2021; 13:E179. [PMID: 33430230 PMCID: PMC7825727 DOI: 10.3390/cancers13020179] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/03/2023] Open
Abstract
Metastatic cancer is one of the major causes of cancer-related mortalities. Metastasis is a complex, multi-process phenomenon, and a hallmark of cancer. Calcium (Ca2+) is a ubiquitous secondary messenger, and it has become evident that Ca2+ signalling plays a vital role in cancer. Ca2+ homeostasis is dysregulated in physiological processes related to tumour metastasis and progression-including cellular adhesion, epithelial-mesenchymal transition, cell migration, motility, and invasion. In this review, we looked at the role of intracellular and extracellular Ca2+ signalling pathways in processes that contribute to metastasis at the local level and also their effects on cancer metastasis globally, as well as at underlying molecular mechanisms and clinical applications. Spatiotemporal Ca2+ homeostasis, in terms of oscillations or waves, is crucial for hindering tumour progression and metastasis. They are a limited number of clinical trials investigating treating patients with advanced stages of various cancer types. Ca2+ signalling may serve as a novel hallmark of cancer due to the versatility of Ca2+ signals in cells, which suggests that the modulation of specific upstream/downstream targets may be a therapeutic approach to treat cancer, particularly in patients with metastatic cancers.
Collapse
Affiliation(s)
- Abeer Alharbi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
- Pharmaceutical Sciences Department, College of Pharmacy, King Saud Bin Abdul-Aziz University for Health Sciences, Riyadh 11426, Saudi Arabia
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK;
| |
Collapse
|
22
|
Wu S, Chen M, Huang J, Zhang F, Lv Z, Jia Y, Cui YZ, Sun LZ, Wang Y, Tang Y, Verhoeft KR, Li Y, Qin Y, Lin X, Guan XY, Lam KO. ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly. Cancer Res 2020; 81:986-1000. [PMID: 33310726 DOI: 10.1158/0008-5472.can-20-0049] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 10/27/2020] [Accepted: 12/07/2020] [Indexed: 11/16/2022]
Abstract
The ubiquitous second messenger Ca2+ has long been recognized as a key regulator in cell migration. Locally confined Ca2+, in particular, is essential for building front-to-rear Ca2+ gradient, which serves to maintain the morphologic polarity required in directionally migrating cells. However, little is known about the source of the Ca2+ and the mechanism by which they crosstalk between different signaling pathways in cancer cells. Here, we report that calcium release-activated calcium modulator 2 (ORAI2), a poorly characterized store-operated calcium (SOC) channel subunit, predominantly upregulated in the lymph node metastasis of gastric cancer, supports cell proliferation and migration. Clinical data reveal that a high frequency of ORAI2-positive cells in gastric cancer tissues significantly correlated with poor differentiation, invasion, lymph node metastasis, and worse prognosis. Gain- and loss-of-function showed that ORAI2 promotes cell motility, tumor formation, and metastasis in both gastric cancer cell lines and mice. Mechanistically, ORAI2 mediated SOC activity and regulated tumorigenic properties through the activation of the PI3K/Akt signaling pathways. Moreover, ORAI2 enhanced the metastatic ability of gastric cancer cells by inducing FAK-mediated MAPK/ERK activation and promoted focal adhesion disassembly at rear-edge of the cell. Collectively, our results demonstrate that ORAI2 is a novel gene that plays an important role in the tumorigenicity and metastasis of gastric cancer. SIGNIFICANCE: These findings describe the critical role of ORAI2 in gastric cancer cell migration and tumor metastasis and uncover the translational potential to advance drug discovery along the ORAI2 signaling pathway.
Collapse
Affiliation(s)
- Shayi Wu
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Miao Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiao Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Feifei Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhaojie Lv
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yongxu Jia
- Department of Clinical Oncology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yu-Zhu Cui
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Liang-Zhan Sun
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Ying Wang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ying Tang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Krista R Verhoeft
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, China
| | - Yanru Qin
- Department of Clinical Oncology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiang Lin
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Ka-On Lam
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
23
|
Pethő Z, Najder K, Carvalho T, McMorrow R, Todesca LM, Rugi M, Bulk E, Chan A, Löwik CWGM, Reshkin SJ, Schwab A. pH-Channeling in Cancer: How pH-Dependence of Cation Channels Shapes Cancer Pathophysiology. Cancers (Basel) 2020; 12:E2484. [PMID: 32887220 PMCID: PMC7565548 DOI: 10.3390/cancers12092484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
Tissue acidosis plays a pivotal role in tumor progression: in particular, interstitial acidosis promotes tumor cell invasion, and is a major contributor to the dysregulation of tumor immunity and tumor stromal cells. The cell membrane and integral membrane proteins commonly act as important sensors and transducers of altered pH. Cell adhesion molecules and cation channels are prominent membrane proteins, the majority of which is regulated by protons. The pathophysiological consequences of proton-sensitive ion channel function in cancer, however, are scarcely considered in the literature. Thus, the main focus of this review is to highlight possible events in tumor progression and tumor immunity where the pH sensitivity of cation channels could be of great importance.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Karolina Najder
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Tiago Carvalho
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
| | - Luca Matteo Todesca
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Micol Rugi
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Etmar Bulk
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| | - Alan Chan
- Percuros B.V., 2333 CL Leiden, The Netherlands;
| | - Clemens W. G. M. Löwik
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3035 GD Rotterdam, The Netherlands; (R.M.); (C.W.G.M.L.)
- Department of Oncology CHUV, UNIL and Ludwig Cancer Center, 1011 Lausanne, Switzerland
| | - Stephan J. Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, 90126 Bari, Italy; (T.C.); (S.J.R.)
| | - Albrecht Schwab
- Institute of Physiology II, University Münster, 48147 Münster, Germany; (K.N.); (L.M.T.); (M.R.); (E.B.); (A.S.)
| |
Collapse
|
24
|
Scremin E, Agostini M, Leparulo A, Pozzan T, Greotti E, Fasolato C. ORAI2 Down-Regulation Potentiates SOCE and Decreases Aβ42 Accumulation in Human Neuroglioma Cells. Int J Mol Sci 2020; 21:ijms21155288. [PMID: 32722509 PMCID: PMC7432374 DOI: 10.3390/ijms21155288] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Senile plaques, the hallmarks of Alzheimer's Disease (AD), are generated by the deposition of amyloid-beta (Aβ), the proteolytic product of amyloid precursor protein (APP), by β and γ-secretase. A large body of evidence points towards a role for Ca2+ imbalances in the pathophysiology of both sporadic and familial forms of AD (FAD). A reduction in store-operated Ca2+ entry (SOCE) is shared by numerous FAD-linked mutations, and SOCE is involved in Aβ accumulation in different model cells. In neurons, both the role and components of SOCE remain quite obscure, whereas in astrocytes, SOCE controls their Ca2+-based excitability and communication to neurons. Glial cells are also directly involved in Aβ production and clearance. Here, we focus on the role of ORAI2, a key SOCE component, in modulating SOCE in the human neuroglioma cell line H4. We show that ORAI2 overexpression reduces both SOCE level and stores Ca2+ content, while ORAI2 downregulation significantly increases SOCE amplitude without affecting store Ca2+ handling. In Aβ-secreting H4-APPswe cells, SOCE inhibition by BTP2 and SOCE augmentation by ORAI2 downregulation respectively increases and decreases Aβ42 accumulation. Based on these findings, we suggest ORAI2 downregulation as a potential tool to rescue defective SOCE in AD, while preventing plaque formation.
Collapse
Affiliation(s)
- Elena Scremin
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
| | - Mario Agostini
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
| | - Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
- Neuroscience Institute—Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35129 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
- Neuroscience Institute—Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Correspondence: (E.G.); (C.F.)
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.S.); (M.A.); (A.L.); (T.P.)
- Correspondence: (E.G.); (C.F.)
| |
Collapse
|
25
|
Abstract
Ca2+ is a ubiquitous and dynamic second messenger molecule that is induced by many factors including receptor activation, environmental factors, and voltage, leading to pleiotropic effects on cell function including changes in migration, metabolism and transcription. As such, it is not surprising that aberrant regulation of Ca2+ signals can lead to pathological phenotypes, including cancer progression. However, given the highly context-specific nature of Ca2+-dependent changes in cell function, delineation of its role in cancer has been a challenge. Herein, we discuss the distinct roles of Ca2+ signaling within and between each type of cancer, including consideration of the potential of therapeutic strategies targeting these signaling pathways.
Collapse
Affiliation(s)
- Scott Gross
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Pranava Mallu
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hinal Joshi
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Bryant Schultz
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Christina Go
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jonathan Soboloff
- Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States; Department of Medical Genetics & Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.
| |
Collapse
|
26
|
Khan HY, Mazahir I, Reddy S, Fazili F, Azmi A. Roles of CRAC channel in cancer: implications for therapeutic development. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:371-382. [PMID: 33728379 DOI: 10.1080/23808993.2020.1803062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction The Ca2+release-activated Ca2+ (CRAC) channel, composed of Orai and STIM proteins, represents one of the main routes of Ca2+ entry in most non-excitable cells. There is accumulating evidence to suggest that CRAC channel can influence various processes associated with tumorigenesis. Overexpression of CRAC channel proteins has been observed in several types of cancer tissues and cells, indicating that blocking CRAC channel activated Ca2+ influx can have therapeutic benefits for cancer patients. Areas covered In this review, we have primarily focused on the molecular composition and activation mechanism of CRAC channel as well as the myriad roles this Ca2+ channel play in various cancers. We further describe relevant information about several efforts aimed at developing CRAC channel blockers and their likely implications for cancer therapy. We have extensively utilized the available literature on PubMed to this end. Expert opinion The possibility of targeting CRAC channel mediated Ca2+ entry in cancer cells has generated considerable interest in recent years. Use of CRAC channel blockers in cancer preclinical studies and clinical trials has been relatively limited as compared to other diseases. The future lies in developing and testing more potent and selective drugs that target cancer cell specific CRAC channel proteins, hence opening better avenues for cancer therapeutic development.
Collapse
Affiliation(s)
- Husain Yar Khan
- Department of Oncology, Wayne State University School of Medicine, Detroit MI 48201 USA
| | - Iqra Mazahir
- Department of Medical Elementology and Toxicology, Jamia Hamdard, Block D, Hamdard Nagar, New Delhi, Delhi 110062, India
| | - Shriya Reddy
- Department of Oncology, Wayne State University School of Medicine, Detroit MI 48201 USA
| | - Farzeen Fazili
- Department of Oncology, Wayne State University School of Medicine, Detroit MI 48201 USA
| | - AsfarSohail Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit MI 48201 USA
| |
Collapse
|
27
|
Lopez JJ, Jardin I, Albarrán L, Sanchez-Collado J, Cantonero C, Salido GM, Smani T, Rosado JA. Molecular Basis and Regulation of Store-Operated Calcium Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:445-469. [PMID: 31646520 DOI: 10.1007/978-3-030-12457-1_17] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous mechanism for Ca2+ influx in mammalian cells with important physiological implications. Since the discovery of SOCE more than three decades ago, the mechanism that communicates the information about the amount of Ca2+ accumulated in the intracellular Ca2+ stores to the plasma membrane channels and the nature of these channels have been matters of intense investigation and debate. The stromal interaction molecule-1 (STIM1) has been identified as the Ca2+ sensor of the intracellular Ca2+ compartments that activates the store-operated channels. STIM1 regulates two types of store-dependent channels: the Ca2+ release-activated Ca2+ (CRAC) channels, formed by Orai1 subunits, that conduct the highly Ca2+ selective current I CRAC and the cation permeable store-operated Ca2+ (SOC) channels, which consist of Orai1 and TRPC1 proteins and conduct the non-selective current I SOC. While the crystal structure of Drosophila CRAC channel has already been solved, the architecture of the SOC channels still remains unclear. The dynamic interaction of STIM1 with the store-operated channels is modulated by a number of proteins that either support the formation of the functional STIM1-channel complex or protect the cell against Ca2+ overload.
Collapse
Affiliation(s)
- Jose J Lopez
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Isaac Jardin
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain.
| | - Letizia Albarrán
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Jose Sanchez-Collado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Carlos Cantonero
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Gines M Salido
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics and Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/University of Sevilla, Sevilla, Spain
| | - Juan A Rosado
- Department of Physiology, Cell Physiology Research Group and Institute of Molecular Pathology Biomarkers, University of Extremadura, Cáceres, Spain
| |
Collapse
|
28
|
Perrouin-Verbe MA, Schoentgen N, Talagas M, Garlantezec R, Uguen A, Doucet L, Rosec S, Marcorelles P, Potier-Cartereau M, Vandier C, Ferec C, Fromont G, Fournier G, Valeri A, Mignen O. Overexpression of certain transient receptor potential and Orai channels in prostate cancer is associated with decreased risk of systemic recurrence after radical prostatectomy. Prostate 2019; 79:1793-1804. [PMID: 31475744 DOI: 10.1002/pros.23904] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Several studies had suggested the potential role of calcium signaling in prostate cancer (PCa) prognosis and agressiveness. We aimed to investigate selected proteins contributing to calcium (Ca2+ ) signaling, (Orai, stromal interaction molecule (STIM), and transient receptor potential (TRP) channels) and involved in cancer hallmarks, as independent predictors of systemic recurrence after radical prostatectomy (RP). METHODS A case-control study including 112 patients with clinically localized PCa treated by RP between 2002 and 2009 and with at least 6-years' follow-up. Patients were divided into two groups according to the absence or presence of systemic recurrence. Expression levels of 10 proteins involved in Ca2+ signaling (TRPC1, TRPC4, TRPV5, TRPV6, TRPM8, STIM1, STIM2, Orai1, Orai2, and Orai3), were assessed by immunohistochemistry using tissue microarrays (TMAs) constructed from paraffin-embedded PCa specimens. The level of expression of the various transcripts in PCa was assessed using quantitative polymerase chain reaction (qPCR) analysis. RNA samples for qPCR were obtained from fresh frozen tissue samples of PCa after laser capture microdissection on RP specimens. Relative gene expression was analyzed using the 2-▵▵Ct method. RESULTS Multivariate analysis showed that increased expression of TRPC1, TRPC4, TRPV5, TRPV6, TRPM8, and Orai2 was significantly associated with a lower risk of systemic recurrence after RP, independently of the prostate-specific antigen (PSA) level, percentage of positive biopsies, and surgical margin (SM) status (P = .007, P = .01, P < .001, P = .0065, P = .007, and P = .01, respectively). For TRPC4, TRPV5, and TRPV6, this association was also independent of Gleason score and pT stage. Moreover, overexpression of TRPV6 and Orai2 was significantly associated with longer time to recurrence after RP (P = .048 and .023, respectively). Overexpression of TRPC4, TRPV5, TRPV6, and Orai2 transcripts was observed in group R- (3.71-, 5.7-, 1.14-, and 2.65-fold increase, respectively). CONCLUSIONS This is the first study to suggest the independent prognostic value of certain proteins involved in Ca2+ influx in systemic recurrence after RP: overexpression of TRPC1, TRPC4, TRPV5, TRPV6, TRPM8, and Orai2 is associated with a lower risk of systemic recurrence. TRPC4, TRPV5, and TRPV6 appear to be particularly interesting, as they are independent of the five commonly used predictive factors, that is, PSA, percentage of positive biopsies, SM status, Gleason score, and pT stage.
Collapse
Affiliation(s)
- M A Perrouin-Verbe
- Department of Urology, CHRU-Université de Brest, Brest, France
- INSERM UMR1078, Université de Bretagne Occidentale, Brest, France
- Department of Urology, CHU-Université de Nantes, Nantes, France
| | - N Schoentgen
- Department of Urology, CHRU-Université de Brest, Brest, France
- INSERM UMR1078, Université de Bretagne Occidentale, Brest, France
| | - M Talagas
- Department of Pathology, CHRU-Université de Brest, Brest, France
- EA 4685 - LIEN, Université de Bretagne Occidentale, Brest, France
| | - R Garlantezec
- INSERM UMR1085-IRSET, Université Rennes 1, Rennes, France
| | - A Uguen
- Department of Pathology, CHRU-Université de Brest, Brest, France
| | - L Doucet
- Department of Pathology, CHRU-Université de Brest, Brest, France
| | - S Rosec
- INSERM UMR1412, Centre d'Investigation Clinique, CHRU-Université de Brest, Brest, France
| | - P Marcorelles
- Department of Pathology, CHRU-Université de Brest, Brest, France
| | | | - C Vandier
- INSERM UMR1069, Université François Rabelais, Tours, France
| | - C Ferec
- INSERM UMR1078, Université de Bretagne Occidentale, Brest, France
| | - G Fromont
- INSERM UMR1069, Université François Rabelais, Tours, France
- Department of Pathology, CHRU-Université de Tours, Tours, France
| | - G Fournier
- Department of Urology, CHRU-Université de Brest, Brest, France
| | - A Valeri
- Department of Urology, CHRU-Université de Brest, Brest, France
| | - O Mignen
- INSERM UMR1078, Université de Bretagne Occidentale, Brest, France
- INSERM UMR1227, Université de Bretagne Occidentale, Brest, France
| |
Collapse
|
29
|
Adenylyl Cyclase Type 8 Overexpression Impairs Phosphorylation-Dependent Orai1 Inactivation and Promotes Migration in MDA-MB-231 Breast Cancer Cells. Cancers (Basel) 2019; 11:cancers11111624. [PMID: 31652779 PMCID: PMC6893434 DOI: 10.3390/cancers11111624] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 12/26/2022] Open
Abstract
Orai1 plays a major role in store-operated Ca2+ entry (SOCE) in triple-negative breast cancer (TNBC) cells. This channel is inactivated via different mechanisms, including protein kinase C (PKC) and protein kinase A (PKA)-dependent phosphorylation at Ser-27 and Ser-30 or Ser-34, respectively, which shapes the Ca2+ responses to agonists. The Ca2+ calmodulin-activated adenylyl cyclase type 8 (AC8) was reported to interact directly with Orai1, thus mediating a dynamic interplay between the Ca2+- and cyclic adenosine monophosphate (cAMP)-dependent signaling pathways. Here, we show that the breast cancer cell lines MCF7 and MDA-MB-231 exhibit enhanced expression of Orai1 and AC8 as compared to the non-tumoral breast epithelial MCF10A cell line. In these cells, AC8 interacts with the Orai1α variant in a manner that is not regulated by Orai1 phosphorylation. AC8 knockdown in MDA-MB-231 cells, using two different small interfering RNAs (siRNAs), attenuates thapsigargin (TG)-induced Ca2+ entry and also Ca2+ influx mediated by co-expression of Orai1 and the Orai1-activating small fragment (OASF) of STIM1 (stromal interaction molecule-1). Conversely, AC8 overexpression enhances SOCE, as well as Ca2+ entry, in cells co-expressing Orai1 and OASF. In MDA-MB-231 cells, we found that AC8 overexpression reduces the Orai1 phosphoserine content, thus suggesting that AC8 interferes with Orai1 serine phosphorylation, which takes place at residues located in the AC8-binding site. Consistent with this, the subset of Orai1 associated with AC8 in naïve MDA-MB-231 cells is not phosphorylated in serine residues in contrast to the AC8-independent Orai1 subset. AC8 expression knockdown attenuates migration of MCF7 and MDA-MB-231 cells, while this maneuver has no effect in the MCF10A cell line, which is likely attributed to the low expression of AC8 in these cells. We found that AC8 is required for FAK (focal adhesion kinase) phosphorylation in MDA-MB-231 cells, which might explain its role in cell migration. Finally, we found that AC8 is required for TNBC cell proliferation. These findings indicate that overexpression of AC8 in breast cancer MDA-MB-231 cells impairs the phosphorylation-dependent Orai1 inactivation, a mechanism that might support the enhanced ability of these cells to migrate.
Collapse
|
30
|
Roberts-Thomson SJ, Chalmers SB, Monteith GR. The Calcium-Signaling Toolkit in Cancer: Remodeling and Targeting. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035204. [PMID: 31088826 DOI: 10.1101/cshperspect.a035204] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Processes that are important in cancer progression, such as sustained cell growth, invasion to other organs, and resistance to cell death inducers, have a clear overlap with pathways regulated by Ca2+ signaling. It is therefore not surprising that proteins important in Ca2+ signaling, sometimes referred to as the "Ca2+ signaling toolkit," can contribute to cancer cell proliferation and invasiveness, and the ability of agents to induce cancer cell death. Ca2+ signaling is also critical in other aspects of cancer progression, including events in the tumor microenvironment and processes involved in the acquisition of resistance to anticancer therapies. This review will consider the role of Ca2+ signaling in tumor progression and highlight areas in which a better understanding of the interplay between the Ca2+-signaling toolkit and tumorigenesis is still required.
Collapse
Affiliation(s)
| | - Silke B Chalmers
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland 4072, Australia.,Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland 4072, Australia
| |
Collapse
|
31
|
Store-Operated Ca 2+ Entry in Tumor Progression: From Molecular Mechanisms to Clinical Implications. Cancers (Basel) 2019; 11:cancers11070899. [PMID: 31252656 PMCID: PMC6678533 DOI: 10.3390/cancers11070899] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 12/18/2022] Open
Abstract
The remodeling of Ca2+ homeostasis has been implicated as a critical event in driving malignant phenotypes, such as tumor cell proliferation, motility, and metastasis. Store-operated Ca2+ entry (SOCE) that is elicited by the depletion of the endoplasmic reticulum (ER) Ca2+ stores constitutes the major Ca2+ influx pathways in most nonexcitable cells. Functional coupling between the plasma membrane Orai channels and ER Ca2+-sensing STIM proteins regulates SOCE activation. Previous studies in the human breast, cervical, and other cancer types have shown the functional significance of STIM/Orai-dependent Ca2+ signals in cancer development and progression. This article reviews the information on the regulatory mechanisms of STIM- and Orai-dependent SOCE pathways in the malignant characteristics of cancer, such as proliferation, resistance, migration, invasion, and metastasis. The recent investigations focusing on the emerging importance of SOCE in the cells of the tumor microenvironment, such as tumor angiogenesis and antitumor immunity, are also reviewed. The clinical implications as cancer therapeutics are discussed.
Collapse
|
32
|
Cantonero C, Sanchez-Collado J, Gonzalez-Nuñez MA, Salido GM, Lopez JJ, Jardin I, Rosado JA. Store-independent Orai1-mediated Ca 2+ entry and cancer. Cell Calcium 2019; 80:1-7. [PMID: 30921687 DOI: 10.1016/j.ceca.2019.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022]
Abstract
Ca2+ channels play an important role in the development of different types of cancer, and considerable progress has been made to understand the pathophysiological mechanisms underlying the role of Ca2+ influx in the development of different cancer hallmarks. Orai1 is among the most ubiquitous and multifunctional Ca2+ channels. Orai1 mediates the highly Ca2+-selective Ca2+ release-activated current (ICRAC) and participates in the less Ca2+-selective store-operated current (ISOC), along with STIM1 or STIM1 and TRPC1, respectively. Furthermore, Orai1 contributes to a variety of store-independent Ca2+ influx mechanisms, including the arachidonate-regulated Ca2+ current, together with Orai3 and the plasma membrane resident pool of STIM1, as well as the constitutive Ca2+ influx processes activated by the secretory pathway Ca2+-ATPase-2 (SPCA2) or supported by physical and functional interaction with the small conductance Ca2+-activated K+ channel 3 (SK3) or the voltage-dependent Kv10.1 channel. This review summarizes the current knowledge concerning the store-independent mechanisms of Ca2+ influx activation through Orai1 channels and their role in the development of different cancer features.
Collapse
Affiliation(s)
- C Cantonero
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J Sanchez-Collado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - M A Gonzalez-Nuñez
- Pathology Service, Hospital San Pedro de Alcantara, 10003 Cáceres, Spain
| | - G M Salido
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J J Lopez
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - I Jardin
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J A Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| |
Collapse
|
33
|
Clemens RA, Lowell CA. CRAC channel regulation of innate immune cells in health and disease. Cell Calcium 2019; 78:56-65. [PMID: 30641250 PMCID: PMC8055042 DOI: 10.1016/j.ceca.2019.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/26/2018] [Accepted: 01/08/2019] [Indexed: 01/17/2023]
Abstract
Calcium is a major intracellular signaling messenger in innate immune cells. Similar to other immune cell subsets, the majority of calcium entry into innate immune cells is induced by cell surface receptors that stimulate store-operated calcium entry through calcium-release activated calcium (CRAC) channels. Since the molecular description of the STIM family of calcium sensors and the ORAI family of CRAC channel proteins, the majority of studies support a dominant role for these proteins in calcium signaling in innate cells. In reviewing the literature on CRAC channel function in innate cells, several general themes emerge. All innate cells express multiple members of the STIM and ORAI family members, however the ratio and relative contribution of individual isoforms changes depending on the cell type and activation state of the cell. It is evident that study of functional roles for STIM molecules is clearly ahead of studies of specific ORAI family members in all innate cell types, and that studies of CRAC channels in innate cells are not nearly as advanced as studies in lymphocytes. However, taken together, evidence from both STIM calcium sensors and ORAI channels in innate cells indicates that deficiency of STIM and ORAI proteins tends not to affect the development of any innate cell lineage, but certainly affects their function, in particular activation of the neutrophil oxidase and mast cell activation via IgE receptors. Furthermore, there are clearly hints that therapeutic targeting of CRAC channels in innate cells offers a new approach to various inflammatory and allergic diseases.
Collapse
Affiliation(s)
- Regina A Clemens
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States.
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California, San Francisco, CA, United States
| |
Collapse
|
34
|
Rode B, Bailey MA, Marthan R, Beech DJ, Guibert C. ORAI Channels as Potential Therapeutic Targets in Pulmonary Hypertension. Physiology (Bethesda) 2019; 33:261-268. [PMID: 29897302 DOI: 10.1152/physiol.00016.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pulmonary hypertension is a complex and fatal disease that lacks treatments. Its pathophysiology involves pulmonary artery hyperreactivity, endothelial dysfunction, wall remodelling, inflammation, and thrombosis, which could all depend on ORAI Ca2+ channels. We review the knowledge about ORAI channels in pulmonary artery and discuss the interest to target them in the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Baptiste Rode
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux , Bordeaux , France.,Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds , Leeds , United Kingdom
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds , Leeds , United Kingdom
| | - Roger Marthan
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux , Bordeaux , France.,Univ. of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux , Bordeaux , France.,CHU de Bordeaux, Pôle Cardio-Thoracique, Bordeaux , France
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds , Leeds , United Kingdom
| | - Christelle Guibert
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux , Bordeaux , France.,Univ. of Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux , Bordeaux , France
| |
Collapse
|
35
|
Immler R, Simon SI, Sperandio M. Calcium signalling and related ion channels in neutrophil recruitment and function. Eur J Clin Invest 2018; 48 Suppl 2:e12964. [PMID: 29873837 PMCID: PMC6221920 DOI: 10.1111/eci.12964] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022]
Abstract
The recruitment of neutrophils to sites of inflammation, their battle against invading microorganisms through phagocytosis and the release of antimicrobial agents is a highly coordinated and tightly regulated process that involves the interplay of many different receptors, ion channels and signalling pathways. Changes in intracellular calcium levels, caused by cytosolic Ca2+ store depletion and the influx of extracellular Ca2+ via ion channels, play a critical role in synchronizing neutrophil activation and function. In this review, we provide an overview of how Ca2+ signalling is initiated in neutrophils and how changes in intracellular Ca2+ levels modulate neutrophil function.
Collapse
Affiliation(s)
- Roland Immler
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Klinikum der Universität, Ludwig-Maximilians-Universität München, Germany
| | - Scott I. Simon
- Department of Biomedical Engineering, Graduate Group in Immunology, University of California, Davis, CA, USA
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Klinikum der Universität, Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
36
|
Diez-Bello R, Jardin I, Lopez JJ, El Haouari M, Ortega-Vidal J, Altarejos J, Salido GM, Salido S, Rosado JA. (-)‑Oleocanthal inhibits proliferation and migration by modulating Ca 2+ entry through TRPC6 in breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:474-485. [PMID: 30321616 DOI: 10.1016/j.bbamcr.2018.10.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/08/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022]
Abstract
Triple negative breast cancer is an aggressive type of cancer that does not respond to hormonal therapy and current therapeutic strategies are accompanied by side effects due to cytotoxic actions on normal tissues. Therefore, there is a need for the identification of anti-cancer compounds with negligible effects on non-tumoral cells. Here we show that (-)‑oleocanthal (OLCT), a phenolic compound isolated from olive oil, selectively impairs MDA-MB-231 cell proliferation and viability without affecting the ability of non-tumoral MCF10A cells to proliferate or their viability. Similarly, OLCT selectively impairs the ability of MDA-MB-231 cells to migrate while the ability of MCF10A to migrate was unaffected. The effect of OLCT was not exclusive for triple negative breast cancer cells as we found that OLCT also attenuate cell viability and proliferation of MCF7 cells. Our results indicate that OLCT is unable to induce Ca2+ mobilization in non-tumoral cells. By contrast, OLCT induces Ca2+ entry in MCF7 and MDA-MB-231 cells, which is impaired by TRPC6 expression silencing. We have found that MDA-MB-231 and MCF7 cells overexpress the channel TRPC6 as compared to non-tumoral MCF10A and treatment with OLCT for 24-72 h downregulates TRPC6 expression in MDA-MB-231 cells. These findings indicate that OLCT impairs the ability of breast cancer cells to proliferate and migrate via downregulation of TRPC6 channel expression while having no effect on the biology of non-tumoral breast cells.
Collapse
Affiliation(s)
- R Diez-Bello
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - I Jardin
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - J J Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - M El Haouari
- Centre Régional des Métiers de l'Education et de la Formation de Taza, 35000 Taza, Morocco; Laboratoire Matériaux, Substances Naturelles, Environnement & Modélisation (LMSNEM), Faculté Polydisciplinaire de Taza, Université Sidi Mohamed Ben Abdellah, Fès, Morocco
| | - J Ortega-Vidal
- Department of Inorganic and Organic Chemistry, University of Jaen, Campus de Excelencia Internacional Agroalimentario (ceiA3), 23071 Jaen, Spain
| | - J Altarejos
- Department of Inorganic and Organic Chemistry, University of Jaen, Campus de Excelencia Internacional Agroalimentario (ceiA3), 23071 Jaen, Spain
| | - G M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
| | - S Salido
- Department of Inorganic and Organic Chemistry, University of Jaen, Campus de Excelencia Internacional Agroalimentario (ceiA3), 23071 Jaen, Spain.
| | - J A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| |
Collapse
|
37
|
Jardin I, Diez-Bello R, Lopez JJ, Redondo PC, Salido GM, Smani T, Rosado JA. TRPC6 Channels Are Required for Proliferation, Migration and Invasion of Breast Cancer Cell Lines by Modulation of Orai1 and Orai3 Surface Exposure. Cancers (Basel) 2018; 10:cancers10090331. [PMID: 30223530 PMCID: PMC6162527 DOI: 10.3390/cancers10090331] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential channels convey signaling information from a number of stimuli to a wide variety of cellular functions, mainly by inducing changes in cytosolic Ca2+ concentration. Different members of the TRPC, TRPM and TRPV subfamilies have been reported to play a role in tumorigenesis. Here we show that the estrogen receptor positive and triple negative breast cancer cell lines, MCF7 and MDA-MB-231, respectively, exhibit enhanced expression of the TRPC6 channel as compared to the non-tumoral MCF10A cell line. In vitro TRPC6 knockdown using shRNA impaired MCF7 and MDA-MB-231 cell proliferation, migration and invasion detected by BrdU incorporation, wound healing and Boyden chamber assays, respectively. Using RNAi-mediated TRPC6 silencing as well as overexpression of the pore-dead dominant-negative TRPC6 mutant we have found that TRPC6 plays a relevant role in the activation of store-operated Ca2+ entry in the breast cancer cell lines but not in non-tumoral breast cells. Finally, we have found that TRPC6 interacts with Orai1 and Orai3 in MCF7 and MDA-MB-231 cells and is required for the translocation of Orai1 and Orai3 to the plasma membrane in MDA-MB-231 and MCF7 cells, respectively, upon Ca2+ store depletion. These findings introduce a novel mechanism for the modulation of Ca2+ influx and the development of different cancer hallmarks in breast cancer cells.
Collapse
Affiliation(s)
- Isaac Jardin
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| | - Raquel Diez-Bello
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| | - Jose J Lopez
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| | - Pedro C Redondo
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| | - Ginés M Salido
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| | - Tarik Smani
- Department of Medical Physiology and Biophysic, Institute of Biomedicine of Sevilla, 41013 Sevilla, Spain.
| | - Juan A Rosado
- Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain.
| |
Collapse
|
38
|
Abstract
Cancer is a major cause of death. The diversity of cancer types and the propensity of cancers to acquire resistance to therapies, including new molecularly targeted and immune-based therapies, drives the search for new ways to understand cancer progression. The remodelling of calcium (Ca2+) signalling and the role of the Ca2+ signal in controlling key events in cancer cells such as proliferation, invasion and the acquisition of resistance to cell death pathways is well established. Most of the work defining such changes has focused on Ca2+ permeable Transient Receptor Potential (TRP) Channels and some voltage gated Ca2+ channels. However, the identification of ORAI channels, a little more than a decade ago, has added a new dimension to how a Ca2+ influx pathway can be remodelled in some cancers and also how calcium signalling could contribute to tumour progression. ORAI Ca2+ channels are now an exemplar for how changes in the expression of specific isoforms of a Ca2+ channel component can occur in cancer, and how such changes can vary between cancer types (e.g. breast cancer versus prostate cancer), and even subtypes (e.g. oestrogen receptor positive versus oestrogen receptor negative breast cancers). ORAI channels and store operated Ca2+ entry are also highlighting the diverse roles of Ca2+ influx pathways in events such as the growth and metastasis of cancers, the development of therapeutic resistance and the contribution of tumour microenvironmental factors in cancer progression. In this review we will highlight some of the studies that have provided evidence for the need to deepen our understanding of ORAI Ca2+ channels in cancer. Many of these studies have also suggested new ways on how we can exploit the role of ORAI channels in cancer relevant processes to develop or inform new therapeutic strategies.
Collapse
|
39
|
Mo P, Yang S. The store-operated calcium channels in cancer metastasis: from cell migration, invasion to metastatic colonization. Front Biosci (Landmark Ed) 2018; 23:1241-1256. [PMID: 28930597 DOI: 10.2741/4641] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Store-operated calcium entry (SOCE) is the predominant calcium entry mechanism in most cancer cells. SOCE is mediated by the endoplasmic reticulum calcium sensor STIMs (STIM1 and 2) and plasma membrane channel forming unit Orais (Orai 1-3). In recent years there is increasing evidence indicating that SOCE in cancer cells is dysregulated to promote cancer cell migration, invasion and metastasis. The overexpression of STIM and Orai proteins has been reported to correlate with the metastatic progression of various cancers. The hyperactive SOCE may promote metastatic dissemination and colonization by reorganizing the actin cytoskeleton, degrading the extracellular matrix and remodeling the tumor microenvironment. Here we discuss how these recent progresses provide novel insights to our understanding of tumor metastasis.
Collapse
Affiliation(s)
- Pingli Mo
- School of Life Sciences, Xiamen University, Xiamen, Fujian China
| | - Shengyu Yang
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033,
| |
Collapse
|
40
|
Kappel S, Marques IJ, Zoni E, Stokłosa P, Peinelt C, Mercader N, Kruithof-de Julio M, Borgström A. Store-Operated Ca 2+ Entry as a Prostate Cancer Biomarker - a Riddle with Perspectives. ACTA ACUST UNITED AC 2017; 3:208-217. [PMID: 29951353 PMCID: PMC6010502 DOI: 10.1007/s40610-017-0072-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose of Review Store-operated calcium entry (SOCE) is dysregulated in prostate cancer, contributing to increased cellular migration and proliferation and preventing cancer cell apoptosis. We here summarize findings on gene expression levels and functions of SOCE components, stromal interaction molecules (STIM1 and STIM2), and members of the Orai protein family (Orai1, 2, and 3) in prostate cancer. Moreover, we introduce new research models that promise to provide insights into whether dysregulated SOCE signaling has clinically relevant implications in terms of increasing the migration and invasion of prostate cancer cells. Recent Findings Recent reports on Orai1 and Orai3 expression levels and function were in part controversial probably due to the heterogeneous nature of prostate cancer. Lately, in prostate cancer cells, transient receptor melastatin 4 channel was shown to alter SOCE and play a role in migration and proliferation. We specifically highlight new cancer research models: a subpopulation of cells that show tumor initiation and metastatic potential in mice and zebrafish models. Summary This review focuses on SOCE component dysregulation in prostate cancer and analyzes several preclinical, cellular, and animal cancer research models.
Collapse
Affiliation(s)
- Sven Kappel
- 1Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland
| | | | - Eugenio Zoni
- 3Urology Research Laboratory, Department of Urology and Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Paulina Stokłosa
- 1Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Christine Peinelt
- 1Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Nadia Mercader
- 2Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- 3Urology Research Laboratory, Department of Urology and Department of Clinical Research, University of Bern, Bern, Switzerland.,4Department of Urology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anna Borgström
- 1Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
41
|
Zhang Y, Li ZY, Hou XX, Wang X, Luo YH, Ying YP, Chen G. Clinical significance and effect of AEG-1 on the proliferation, invasion, and migration of NSCLC: a study based on immunohistochemistry, TCGA, bioinformatics, in vitro and in vivo verification. Oncotarget 2017; 8:16531-16552. [PMID: 28152520 PMCID: PMC5369983 DOI: 10.18632/oncotarget.14972] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Background Astrocyte elevated gene-1 (AEG-1) is related to the tumorigenesis and deterioration of different cancers, including non-small cell lung cancer (NSCLC). However, the effect of AEG-1 in NSCLC remains unclear. In this study, we aimed to investigate the clinical significance and effect of AEG-1 on biological function of NSCLC. Results AEG-1 was significantly overexpressed in NSCLC tissues and closely correlated to the deterioration of NSCLC based on tissue microarray, TCGA database and meta-analysis. After knock-down of AEG-1, the proliferation, migration and invasion of NSCLC cells were all inhibited, and the tumorigenic and angiogenic ability of NSCLC cells were weakened. Furthermore, the AEG-1 co-expressed genes were significantly related to AMPK signaling pathway based on bioinformatics approaches. Materials and Methods A tissue microarray, the Cancer Genome Atlas (TCGA) database, as well as a meta-analysis were performed to analyze the relationship between AEG-1 and the clinicopathological parameters of NSCLC. Furthermore, immunocytochemistry, Western blot analysis, scratch assay, colony formation assay, Transwell migration and invasion assay and the chick embryo chorioallantoic membrane (CAM) model were conducted to explore the effect of AEG-1 on NSCLC in vitro and in vivo. Additionally, bioinformatics analyses were carried out to assess the potential pathways and networks of the co-expressed genes of AEG-1. Conclusions AEG-1 is positively activated in the tumorigenesis and deterioration of NSCLC. We hypothesize that AEG-1 could play an important role in NSCLC via AMPK signaling pathway. Inhibiting the expression of AEG-1 is expected to become a novel method in the therapeutic strategies of NSCLC.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Zu-Yun Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Xin-Xi Hou
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Xiao Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yi-Huan Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Yan-Ping Ying
- Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530021, China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| |
Collapse
|
42
|
Niemeyer BA. The STIM-Orai Pathway: Regulation of STIM and Orai by Thiol Modifications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:99-116. [PMID: 28900911 DOI: 10.1007/978-3-319-57732-6_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cysteines are among the least abundant amino acids found in proteins. Due to their unique nucleophilic thiol group, they are able to undergo a broad range of chemical modifications besides their known role in disulfide formation, such as S-sulfenylation (-SOH), S-sulfinylation (-SO(2)H), S-sufonylation (-SO(3)H), S-glutathionylation (-SSG), and S-sulfhydration (-SSH), among others. These posttranslational modifications can be irreversible and act as transitional modifiers or as reversible on-off switches for the function of proteins. Disturbances of the redox homeostasis, for example, in situations of increased oxidative stress, can contribute to a range of diseases. Because Ca2+ signaling mediated by store-operated calcium entry (SOCE) is involved in a plethora of cellular responses, the cross-talk between reactive oxygen species (ROS) and Ca2+ is critical for homeostatic control. Identification of calcium regulatory protein targets of thiol redox modifications is needed to understand their role in biology and disease.
Collapse
Affiliation(s)
- Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany.
| |
Collapse
|