1
|
Macias-Díaz A, Nieto-Felipe J, Jardín I, Camello PJ, Martinez-Quintana EM, Salido GM, Smani T, Lopez JJ, Rosado JA. Filamin A C-terminal fragment modulates Orai1 expression by inhibition of protein degradation. Am J Physiol Cell Physiol 2025; 328:C657-C669. [PMID: 39764579 DOI: 10.1152/ajpcell.00745.2024] [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: 10/08/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025]
Abstract
Filamin A (FLNA) is an actin-binding protein that has been reported to interact with STIM1 modulating the activation of Orai1 channels. Cleaving of FLNA by calpain leads to a C-terminal fragment that is involved in a variety of functional and pathological events, including pro-oncogenic activity in different types of cancer. Here, we show that full-length FLNA is downregulated in samples from patients with colon cancer as well as in the adenocarcinoma cell line HT-29. This is consistent with an increased calpain-dependent FLNA cleaving with enhanced expression of the C-terminal FLNA fragment accompanied by enhanced expression of Orai1 and STIM1, as well as store-operated Ca2+ entry (SOCE). To further explore the mechanism underlying the enhancement of SOCE by the C-terminal FLNA fragment, we expressed in HEK-293 cells the C-terminal FLNA region encompassing repeats 16-24 (FLNA16-24 fragment), which enhanced both Orai1 and STIM1 as well as SOCE. Transfection of the FLNA16-24 fragment attenuates Orai1 and STIM1 protein degradation, and, specifically, abrogates Orai1α lysosomal degradation and retains this channel in the plasma membrane. However, the C-terminal FLNA fragment did not induce a detectable modification in Orai1β degradation. Due to the relevance of SOCE in cell physiology, our results provide evidence of a novel mechanism for the regulation of Ca2+ influx with relevant pathophysiological implications.NOTE & NOTEWORTHY FLNA cleaving by calpain has been observed in a variety of tumoral, including prostate and colorectal cancer cells, as well as in nontumoral cells, leading to a C-terminal fragment encompassing repeats 16-24. Expression of the FLNA16-24 fragment in HEK-293 cells enhances Orai1 and STIM1 expression, as well as SOCE, a mechanism mediated by attenuation of Orai1α and STIM1 degradation, providing evidence for a novel mechanism for the regulation of SOCE in normal and malignant cells.
Collapse
Affiliation(s)
- Alvaro Macias-Díaz
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Joel Nieto-Felipe
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Pedro J Camello
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | | | - Gines M Salido
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Tarik Smani
- Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Seville, Spain
- Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, Seville, Spain
| | - Jose J Lopez
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology (Cellular Physiology Research Group), Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, Caceres, Spain
| |
Collapse
|
2
|
Serano M, Perni S, Pierantozzi E, Laurino A, Sorrentino V, Rossi D. Intracellular Membrane Contact Sites in Skeletal Muscle Cells. MEMBRANES 2025; 15:29. [PMID: 39852269 PMCID: PMC11767089 DOI: 10.3390/membranes15010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025]
Abstract
Intracellular organelles are common to eukaryotic cells and provide physical support for the assembly of specialized compartments. In skeletal muscle fibers, the largest intracellular organelle is the sarcoplasmic reticulum, a specialized form of the endoplasmic reticulum primarily devoted to Ca2+ storage and release for muscle contraction. Occupying about 10% of the total cell volume, the sarcoplasmic reticulum forms multiple membrane contact sites, some of which are unique to skeletal muscle. These contact sites primarily involve the plasma membrane; among these, specialized membrane contact sites between the transverse tubules and the terminal cisternae of the sarcoplasmic reticulum form triads. Triads are skeletal muscle-specific contact sites where Ca2+ channels and regulatory proteins assemble to form the so-called calcium release complex. Additionally, the sarcoplasmic reticulum contacts mitochondria to enable a more precise regulation of Ca2+ homeostasis and energy metabolism. The sarcoplasmic reticulum and the plasma membrane also undergo dynamic remodeling to allow Ca2+ entry from the extracellular space and replenish the stores. This process involves the formation of dynamic membrane contact sites called Ca2+ Entry Units. This review explores the key processes in biogenesis and assembly of intracellular membrane contact sites as well as the membrane remodeling that occurs in response to muscle fatigue.
Collapse
Affiliation(s)
- Matteo Serano
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Stefano Perni
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Annunziatina Laurino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (M.S.); (S.P.); (E.P.); (A.L.); (V.S.)
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| |
Collapse
|
3
|
Nieto‐Felipe J, Macias‐Díaz A, Jimenez‐Velarde V, Lopez JJ, Salido GM, Smani T, Jardin I, Rosado JA. Feedback modulation of Orai1α and Orai1β protein content mediated by STIM proteins. J Cell Physiol 2025; 240:e31450. [PMID: 39359018 PMCID: PMC11730744 DOI: 10.1002/jcp.31450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/03/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
Store-operated Ca2+ entry is a mechanism controlled by the filling state of the intracellular Ca2+ stores, predominantly the endoplasmic reticulum (ER), where ER-resident proteins STIM1 and STIM2 orchestrate the activation of Orai channels in the plasma membrane, and Orai1 playing a predominant role. Two forms of Orai1, Orai1α and Orai1β, have been identified, which arises the question whether they are equally regulated by STIM proteins. We demonstrate that STIM1 preferentially activates Orai1α over STIM2, yet both STIM proteins similarly activate Orai1β. Under resting conditions, there is a pronounced association between STIM2 and Orai1α. STIM1 and STIM2 are also shown to influence the protein levels of the Orai1 variants, independently of Ca2+ influx, via lysosomal degradation. Interestingly, Orai1α and Orai1β appear to selectively regulate the protein level of STIM1, but not STIM2. These observations offer crucial insights into the regulatory dynamics between STIM proteins and Orai1 variants, enhancing our understanding of the intricate processes that fine-tune intracellular Ca2+ signaling.
Collapse
Affiliation(s)
- Joel Nieto‐Felipe
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Alvaro Macias‐Díaz
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Vanesa Jimenez‐Velarde
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Jose J. Lopez
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Gines M. Salido
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Tarik Smani
- Group of Cardiovascular PathophysiologyInstitute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSICSevilleSpain
- Department of Medical Physiology and BiophysicsFaculty of Medicine, University of SevilleSevilleSpain
| | - Isaac Jardin
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| | - Juan A. Rosado
- Department of Physiology (Cellular Physiology Research Group)Institute of Molecular Pathology Biomarkers (IMPB), University of ExtremaduraCaceresSpain
| |
Collapse
|
4
|
Gomaa AAE, Zeid AMA, Nagy IM, Zahran AM. The effect of genetic polymorphisms in STIM1 and ORAI1 on erythropoietin resistance in Egyptian patients with end-stage renal disease. Clin Chim Acta 2025; 564:119948. [PMID: 39214396 DOI: 10.1016/j.cca.2024.119948] [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: 08/02/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Chronic renal failure (CRF) is an incurable disease with unique challenges. Anemia is a frequent complication affecting dialysis patients. Erythropoietin (EPO) is used to treat anemia, but a poor response may result. We investigated genetic polymorphisms of store-operated calcium channel (SOC) signaling, an important erythropoietin-activated pathway that may induce EPO resistance in patients with renal failure. A total of 108 end stage renal disease (ESRD) patients were selected for this study. Patients were divided into two groups according to their erythropoietin resistance index (ERI): 39 patients with an ERI>10 and 69 patients with an ERI<10. We selected four tagging single nucleotide polymorphisms (tSNPs) in STIM1 and five in ORAI1 in our study. A polymerase chain reaction was performed, and genotyping against EPO resistance was correlated. Patients with the AG genotype of rs1561876 in STIM1, the TC genotype of rs6486795 in ORAI1, and the TG or GG genotypes of rs12320939 in ORAI1 were associated with an increased risk of erythropoietin resistance. Overall, we reported a moderately significant relationship between genetic polymorphisms of STIM1 and EPO resistance. We also reported a highly significant relationship between genetic polymorphisms of ORAI1 and EPO resistance. The (A-A-G) haplotype of STIM1 and the (G-T-G-T-A, G-C-G-C-G, or G-T-T-C-G) haplotypes of ORAI1 were significantly associated with EPO resistance.
Collapse
Affiliation(s)
- Azza A E Gomaa
- Internal Medicine Department, Menofia University, Menofia, Egypt.
| | - Amany M A Zeid
- Clinical Pathology Department, Menofia University, Menofia, Egypt
| | - Ibrahim M Nagy
- Medicinal Chemistry Department, Menofia University, Menofia, Egypt.
| | - Ahmed M Zahran
- Internal Medicine Department, Menofia University, Menofia, Egypt
| |
Collapse
|
5
|
Chowdhury D, Jang CE, Lajoie P, Renaud SJ. A stress paradox: the dual role of the unfolded protein response in the placenta. Front Endocrinol (Lausanne) 2024; 15:1525189. [PMID: 39758342 PMCID: PMC11695235 DOI: 10.3389/fendo.2024.1525189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
The placenta is a temporary organ that forms during pregnancy and is essential for fetal development and maternal health. As an endocrine organ, proper placental function requires continual production, folding, and transport of proteins and lipids. Central to these processes is the endoplasmic reticulum (ER), a dynamic organelle responsible for maintaining cellular protein and lipid synthesis and processing. ER stress occurs when there is an accumulation of unfolded or misfolded proteins, which triggers the activation of cellular pathways collectively called the unfolded protein response. Unfolded protein response pathways act to alleviate the misfolded protein burden and restore ER homeostasis, or if unresolved, initiate cell death. While prolonged ER stress has been linked to deficient placental function and adverse pregnancy outcomes, basal activation of unfolded protein response pathways is required for placental development and function. This review explores the importance of ER homeostasis in placental development and function, examining how disruptions in ER stress responses may contribute to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Diba Chowdhury
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Chloe E. Jang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| | - Stephen J. Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
6
|
Pandrangi SL, Chittineedi P, Manthari RK, Suhruth B. Impact of oxytosis on the cross-talk of mTORC with mitochondrial proteins in drug-resistant cancer stem cells. J Cell Physiol 2024; 239:e31421. [PMID: 39188055 PMCID: PMC11649969 DOI: 10.1002/jcp.31421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
By delivering the environmental inputs to transport nutrients and growth factors, Mechanistic Target of Rapamycin (mTOR) plays a significant role in the growth and metabolism of eukaryotic cells through the regulation of numerous elementary cellular processes such as autophagy, protein synthesis, via translation of mitochondrial protein transcription factor A mitochondrial, mitochondrial ribosomal proteins, and mitochondrial respiratory complexes I &V that are encoded in the nucleus with the help of translation initiation factor 4E-BP. These mitochondrial proteins are involved in cell signaling to regulate proper cell growth, proliferation, and death which are essential for tumor growth and proliferation. This suggests that tumor cells are dependent on mTORC1 for various metabolic pathways. However, this crucial regulator is activated and regulated by calcium homeostasis. Mounting evidence suggests the role of calcium ions in regulating mitochondrial enzymes and proteins. Hence, disrupting calcium homeostasis leads to calcium-dependent cell death called "Oxytosis" through hampering the expression of various mitochondrial proteins. "Oxytosis" is a novel non-apoptotic cell death characterized by glutamate cytotoxicity and ferritin degradation. The present review focuses on the crosstalk between mTORC1 and mitochondrial proteins in the cancer pathophysiology and the impact of calcium ions on disrupting mTORC1 leading to the induction of "Oxytosis."
Collapse
Affiliation(s)
- Santhi L. Pandrangi
- Department of Life Sciences, School of ScienceGITAM (Deemed to be) UniversityVisakhapatnamIndia
| | - Prasanthi Chittineedi
- Department of Life Sciences, School of ScienceGITAM (Deemed to be) UniversityVisakhapatnamIndia
| | - Ram K. Manthari
- Department of Life Sciences, School of ScienceGITAM (Deemed to be) UniversityVisakhapatnamIndia
| | - Balaji Suhruth
- Department of Life Sciences, School of ScienceGITAM (Deemed to be) UniversityVisakhapatnamIndia
| |
Collapse
|
7
|
Zhao N, Michelucci A, Pietrangelo L, Malik S, Groom L, Leigh J, O'Connor TN, Takano T, Kingsley PD, Palis J, Boncompagni S, Protasi F, Dirksen RT. An Orai1 gain-of-function tubular aggregate myopathy mouse model phenocopies key features of the human disease. EMBO J 2024; 43:5941-5971. [PMID: 39420094 PMCID: PMC11612304 DOI: 10.1038/s44318-024-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Tubular aggregate myopathy (TAM) is a heritable myopathy primarily characterized by progressive muscle weakness, elevated levels of creatine kinase (CK), hypocalcemia, exercise intolerance, and the presence of tubular aggregates (TAs). Here, we generated a knock-in mouse model based on a human gain-of-function mutation which results in a severe, early-onset form of TAM, by inducing a glycine-to-serine point mutation in the ORAI1 pore (Orai1G100S/+ or GS mice). By 8 months of age, GS mice exhibited significant muscle weakness, exercise intolerance, elevated CK levels, hypocalcemia, and robust TA presence. Unexpectedly, constitutive Ca2+ entry in mutant mice was observed in muscle only during early development and was abolished in adult skeletal muscle, partly due to reduced ORAI1 expression. Consistent with proteomic results, significant mitochondrial damage and dysfunction was observed in skeletal muscle of GS mice. Thus, GS mice represent a powerful model for investigation of the pathophysiological mechanisms that underlie key TAM symptoms, as well as those compensatory responses that limit the damaging effects of uncontrolled ORAI1-mediated Ca2+ influx.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Antonio Michelucci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Linda Groom
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas N O'Connor
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience and Clinical Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology & DMSI, Department of Medicine and Aging Sciences, University Gabriele d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
8
|
Makio T, Chen J, Simmen T. ER stress as a sentinel mechanism for ER Ca 2+ homeostasis. Cell Calcium 2024; 124:102961. [PMID: 39471738 DOI: 10.1016/j.ceca.2024.102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Endoplasmic reticulum (ER) stress is triggered upon the interference with oxidative protein folding that aims to produce fully folded, disulfide-bonded and glycosylated proteins, which are then competent to exit the ER. Many of the enzymes catalyzing this process require the binding of Ca2+ ions, including the chaperones BiP/GRP78, calnexin and calreticulin. The induction of ER stress with a variety of drugs interferes with chaperone Ca2+ binding, increases cytosolic Ca2+through the opening of ER Ca2+ channels, and activates store-operated Ca2+ entry (SOCE). Posttranslational modifications (PTMs) of the ER Ca2+ handling proteins through ER stress-dependent phosphorylation or oxidation control these mechanisms, as demonstrated in the case of the sarco/endoplasmic reticulum ATPase (SERCA), inositol 1,4,5 trisphosphate receptors (IP3Rs) or stromal interaction molecule 1 (STIM1). Their aim is to restore ER Ca2+ homeostasis but also to increase Ca2+ transfer from the ER to mitochondria during ER stress. This latter function boosts ER bioenergetics, but also triggers apoptosis if ER Ca2+ signaling persists. ER Ca2+ toolkit oxidative modifications upon ER stress can occur within the ER lumen or in the adjacent cytosol. Enzymes involved in this redox control include ER oxidoreductin 1 (ERO1) or the thioredoxin-family protein disulfide isomerases (PDI) and ERp57. A tight, but adaptive connection between ER Ca2+ content, ER stress and mitochondrial readouts allows for the proper functioning of many tissues, including skeletal muscle, the liver, and the pancreas, where ER stress either maintains or compromises their function, depending on its extent and context. Upon mutation of key regulators of ER Ca2+ signaling, diseases such as muscular defects (e.g., from mutated selenoprotein N, SEPN1/SELENON), or diabetes (e.g., from mutated PERK) are the result.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada.
| |
Collapse
|
9
|
Lu X, Wang Y, Yu K, Li M, Yang X, Shen Y. Activated human Orai1 channel in lipid biolayer may exist as a pentamer. Biochem Biophys Res Commun 2024; 733:150723. [PMID: 39312878 DOI: 10.1016/j.bbrc.2024.150723] [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: 09/18/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024]
Abstract
The human Orai1 (hOrai1) channel plays a crucial role in extracellular Ca2+ influx and has emerged as an attractive drug target for various diseases. However, the activated structure of the hOrai1 channel assembly within a lipid bilayer remains unknown. In this study, we expressed and purified the hOrai1 channel covalently linked to two SOAR tandems (HOSS). Patch-clamp experiments in whole-cell configuration showed that HOSS is constitutively active. Biochemical characterization confirmed that the purified HOSS channels were successfully incorporated into MSP1E3D1 nanodiscs. Negative staining revealed that the HOSS channels resemble a mushroom, with the body representing the hOrai1 channel and the leg representing the SOAR domain. Surprisingly, 2D analysis of cryo-EM data demonstrated a pentameric assembly of HOSS in a lipid bilayer. Our findings suggest that the hOrai1 channel may assemble into different oligomeric states in response to varying membrane environments.
Collapse
Affiliation(s)
- Xuhang Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yaojie Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Keer Yu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Ming Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xue Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Yuequan Shen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
10
|
Tripoli BA, Smyth JT. Septins regulate heart contractility through modulation of cardiomyocyte store-operated calcium entry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621876. [PMID: 39574715 PMCID: PMC11580947 DOI: 10.1101/2024.11.04.621876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Highly regulated cardiomyocyte Ca 2+ fluxes drive heart contractions. Recent findings from multiple organisms demonstrate that the specific Ca 2+ transport mechanism known as store-operated Ca 2+ entry (SOCE) is essential in cardiomyocytes for proper heart function, and SOCE dysregulation results in cardiomyopathy. Mechanisms that regulate SOCE in cardiomyocytes are poorly understood. Here we tested the role of cytoskeletal septin proteins in cardiomyocyte SOCE regulation. Septins are essential SOCE modulators in other cell types, but septin functions in cardiomyocytes are nearly completely unexplored. We show using targeted genetics and intravital imaging of heart contractility in Drosophila that cardiomyocyte-specific depletion of septins 1, 2, and 4 results in heart dilation that phenocopies the effects of SOCE suppression. Heart dilation caused by septin 2 depletion was suppressed by SOCE upregulation, supporting the hypothesis that septin 2 is required in cardiomyocytes for sufficient SOCE function. A major function of SOCE is to support SERCA-dependent sarco/endoplasmic reticulum (S/ER) Ca 2+ stores, and augmenting S/ER store filling by SERCA overexpression also suppressed the septin 2 phenotype. We also ruled out several potential SOCE-independent septin functions, as septin 2 phenotypes were not due to septin function during development and septin 2 was not required for z-disk organization as defined by α-actinin labeling. These results demonstrate, for the first time, an essential role of septins in cardiomyocyte physiology and heart function that is due, at least in part, to septin regulation of SOCE function.
Collapse
|
11
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Plasma membrane curvature regulates the formation of contacts with the endoplasmic reticulum. Nat Cell Biol 2024; 26:1878-1891. [PMID: 39289582 PMCID: PMC11567891 DOI: 10.1038/s41556-024-01511-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules, we hypothesize that PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, whereas the ubiquitously expressed extended synaptotagmin-2 does not show a preference for PM curvature. At the mechanistic level, we find that the low-complexity region (LCR) and membrane occupation and recognition nexus (MORN) motifs of junctophilins can bind independently to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins-Eps15 homology domain-containing proteins-that interact with the MORN_LCR motifs and facilitate the preferential tethering of junctophilins to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Luis A Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Departments of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical and Nano Engineering, University of California, San Diego, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples, Italy
- Faculty of Electrical Engineering and Information Technology, RWTH Aachen University, Aachen, Germany
- Institute of Biological Information Processing-Bioelectronics (IBI-3), Forschungszentrum, Jülich, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
12
|
Luo X, Jian M, Wu P, Wu Y, Ma Y, Feng N, Lu M, Shi D, Liu R, Ding Y, Zhang W, Fan L, He X. STIM1 promotes cervical cancer progression through autophagy activation via TFEB nuclear translocation. Cell Signal 2024; 125:111500. [PMID: 39489201 DOI: 10.1016/j.cellsig.2024.111500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/13/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Autophagy plays an important role in maintaining the stability of intracellular environment, abnormal autophagy is associated with the occurrence and progression of cancer, the role of STIM1 in regulating cancer autophagy remains controversial, and its clinical relevance is unclear. Our study aimed to investigate the effect and mechanism of STIM1 on cervical cancer, thus to provide new molecular therapeutic targets for cervical cancer in clinic. METHODS We collected CIN III, FIGO IB and IIA fresh Specimens without chemotherapy from patients in Renmin Hospital of Hubei University of Medicine (n = 10). STIM1, TFEB and autophagy related proteins of different stage tissues were detected. In vitro, SKF96365 and AncoA4 were used to inhibit STIM1-administrated Ca2+ entry of SiHa cells, Cyclosporine A (calcineurin inhibitors) were used to inhibit CaN/TFEB pathway, Ad-mCherry-GFPLC3B was used to detect autophagy flux, shSTIM1 was used to knockdown STIM1 expression. RESULTS The expression levels of STIM1, TFEB and autophagy related proteins were positively correlated with the progression of cervical cancer. Inhibition of STIM1-mediated SOCE can decrease proliferation and migration, and promoted the apoptosis of cervical cancer cells. Knockdown STIM1 can inhibit autophagy and TFEB nuclear translocation. CONCLUSION STIM1 can promote autophagy and accelerate cervical cancer progression by increasing TFEB nuclear translocation of cervical cancer cells.
Collapse
Affiliation(s)
- Xi Luo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China; Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Mengchan Jian
- Department of Gynecology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ping Wu
- Department of Reproductive Medicine, Shenzhen Luohu People's Hospital, Shenzhen 518000, China
| | - Yahua Wu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Yulan Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Na Feng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Min Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Dandan Shi
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Rui Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China
| | - Wenjun Zhang
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Li Fan
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Xiju He
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan 442000, China; Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.
| |
Collapse
|
13
|
Al-Ali H, Baig A, Alkhanjari RR, Murtaza ZF, Alhajeri MM, Elbahrawi R, Abdukadir A, Bhamidimarri PM, Kashir J, Hamdan H. Septins as key players in spermatogenesis, fertilisation and pre-implantation embryogenic cytoplasmic dynamics. Cell Commun Signal 2024; 22:523. [PMID: 39468561 PMCID: PMC11514797 DOI: 10.1186/s12964-024-01889-z] [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: 08/22/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Septins are a family of cytokinesis-related proteins involved in regulating cytoskeletal design, cell morphology, and tissue morphogenesis. Apart from cytokinesis, as a fourth component of cytoskeleton, septins aid in forming scaffolds, vesicle sorting and membrane stability. They are also known to be involved in the regulation of intracellular calcium (Ca2+) via the STIM/Orai complex. Infertility affects ~ 15% of couples globally, while male infertility affects ~ 7% of men. Global pregnancy and live birth rates following fertility treatment remain relatively low, while there has been an observable decline in male fertility parameters over the past 60 years. Low fertility treatment success can be attributed to poor embryonic development, poor sperm parameters and fertilisation defects. While studies from the past few years have provided evidence for the role of septins in fertility related processes, the functional role of septins and its related complexes in cellular processes such as oocyte activation, fertilization, and sperm maturation are not completely understood. This review summarizes the available knowledge on the role of septins in spermatogenesis and oocyte activation via Ca2+ regulation, and cytoskeletal dynamics throughout pre-implantation embryonic development. We aim to identify the currently less known mechanisms by which septins regulate these immensely important mechanisms with a view of identifying areas of investigation that would benefit our understanding of cell and reproductive biology, but also provide potential avenues to improve current methods of fertility treatment.
Collapse
Affiliation(s)
- Hana Al-Ali
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Amna Baig
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Rayyah R Alkhanjari
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Zoha F Murtaza
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Maitha M Alhajeri
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Rawdah Elbahrawi
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Azhar Abdukadir
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Poorna Manasa Bhamidimarri
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates
| | - Junaid Kashir
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates.
- Center for Biotechnology, Khalifa University, 127788, Abu Dhabi, United Arab Emirates.
| | - Hamdan Hamdan
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, 127788, United Arab Emirates.
| |
Collapse
|
14
|
Soll D, Chu CF, Sun S, Lutz V, Arunkumar M, Gachechiladze M, Schäuble S, Alissa-Alkhalaf M, Nguyen T, Khalil MA, Garcia-Ribelles I, Mueller M, Buder K, Michalke B, Panagiotou G, Ziegler-Martin K, Benz P, Schatzlmaier P, Hiller K, Stockinger H, Luu M, Schober K, Moosmann C, Schamel WW, Huber M, Zielinski CE. Sodium chloride in the tumor microenvironment enhances T cell metabolic fitness and cytotoxicity. Nat Immunol 2024; 25:1830-1844. [PMID: 39198632 PMCID: PMC11436365 DOI: 10.1038/s41590-024-01918-6] [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] [Accepted: 07/04/2024] [Indexed: 09/01/2024]
Abstract
The efficacy of antitumor immunity is associated with the metabolic state of cytotoxic T cells, which is sensitive to the tumor microenvironment. Whether ionic signals affect adaptive antitumor immune responses is unclear. In the present study, we show that there is an enrichment of sodium in solid tumors from patients with breast cancer. Sodium chloride (NaCl) enhances the activation state and effector functions of human CD8+ T cells, which is associated with enhanced metabolic fitness. These NaCl-induced effects translate into increased tumor cell killing in vitro and in vivo. Mechanistically, NaCl-induced changes in CD8+ T cells are linked to sodium-induced upregulation of Na+/K+-ATPase activity, followed by membrane hyperpolarization, which magnifies the electromotive force for T cell receptor (TCR)-induced calcium influx and downstream TCR signaling. We therefore propose that NaCl is a positive regulator of acute antitumor immunity that might be modulated for ex vivo conditioning of therapeutic T cells, such as CAR T cells.
Collapse
Affiliation(s)
| | - Chang-Feng Chu
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Shan Sun
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Mahima Arunkumar
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Mariam Gachechiladze
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Maha Alissa-Alkhalaf
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Trang Nguyen
- Institute of Biology III, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Michelle-Amirah Khalil
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Ignacio Garcia-Ribelles
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Michael Mueller
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | | | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich-German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Kai Ziegler-Martin
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Pascal Benz
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Philipp Schatzlmaier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Maik Luu
- Chair for Cellular Immunotherapy, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Kilian Schober
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Carolin Moosmann
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang W Schamel
- Institute of Biology III, Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency, University Clinics and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps-University Marburg, Marburg, Germany
| | - Christina E Zielinski
- Technical University of Munich, Munich, Germany.
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany.
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
15
|
Stern MA, Cole ER, Gutekunst CA, Yang JJ, Berglund K, Gross RE. Organellular imaging in vivo reveals a depletion of endoplasmic reticular calcium during post-ictal cortical spreading depolarization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614252. [PMID: 39386598 PMCID: PMC11463492 DOI: 10.1101/2024.09.21.614252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
During cortical spreading depolarization (CSD), neurons exhibit a dramatic increase in cytosolic calcium, which may be integral to CSD-mediated seizure termination. This calcium increase greatly exceeds that during seizures, suggesting the calcium source may not be solely extracellular. Thus, we sought to determine if the endoplasmic reticulum (ER), the largest intracellular calcium store, is involved. We developed a two-photon calcium imaging paradigm to simultaneously record the cytosol and ER during seizures in awake mice. Paired with direct current recording, we reveal that CSD can manifest as a slow post-ictal cytosolic calcium wave with a concomitant depletion of ER calcium that is spatiotemporally consistent with a calcium-induced calcium release. Importantly, we observed both naturally occurring and electrically induced CSD suppressed post-ictal epileptiform activity. Collectively, this work links ER dynamics to CSD, which serves as an innate process for seizure suppression and a potential mechanism underlying therapeutic electrical stimulation for epilepsy.
Collapse
Affiliation(s)
- Matthew A. Stern
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Eric R. Cole
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
- Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
| | - Claire-Anne Gutekunst
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, United States
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Robert E. Gross
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
16
|
Li Y, Zhou L, Deng H, Zhang Y, Li G, Yu H, Wu K, Wang F. A switch in the pathway of TRPC3-mediated calcium influx into brain pericytes contributes to capillary spasms after subarachnoid hemorrhage. Neurotherapeutics 2024; 21:e00380. [PMID: 38839450 PMCID: PMC11581875 DOI: 10.1016/j.neurot.2024.e00380] [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: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024] Open
Abstract
Calcium influx and subsequent elevation of the intracellular calcium concentration ([Ca2+]i) induce contractions of brain pericytes and capillary spasms following subarachnoid hemorrhage. This calcium influx is exerted through cation channels. However, the specific calcium influx pathways in brain pericytes after subarachnoid hemorrhage remain unknown. Transient receptor potential canonical 3 (TRPC3) is the most abundant cation channel potentially involved in calcium influx into brain pericytes and is involved in calcium influx into other cell types either via store-operated calcium entry (SOCE) or receptor-operated calcium entry (ROCE). Therefore, we hypothesized that TRPC3 is associated with [Ca2+]i elevation in brain pericytes, potentially mediating brain pericyte contraction and capillary spasms after subarachnoid hemorrhage. In this study, we isolated rat brain pericytes and demonstrated increased TRPC3 expression and its currents in brain pericytes after subarachnoid hemorrhage. Calcium imaging of brain pericytes revealed that changes in TRPC3 expression mediated a switch from SOCE-dominant to ROCE-dominant calcium influx after subarachnoid hemorrhage, resulting in significantly higher [Ca2+]i levels after SAH. TRPC3 activity in brain pericytes also contributed to capillary spasms and reduction in cerebral blood flow in an in vivo rat model of subarachnoid hemorrhage. Therefore, we suggest that the switch in TRPC3-mediated calcium influx pathways plays a crucial role in the [Ca2+]i elevation in brain pericytes after subarachnoid hemorrhage, ultimately leading to capillary spasms and a reduction in cerebral blood flow.
Collapse
Affiliation(s)
- Yuncong Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Lei Zhou
- The Key Laboratory of Stem Cell and Regenerative Medicine of Yunnan Province, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming 650500, China
| | - Hongji Deng
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yongjin Zhang
- Department of Laboratory for Basic Research, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Guibo Li
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Hanfu Yu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kun Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
17
|
Zhuang Z, Meng Y, Xue Y, Wang Y, Cheng X, Jing J. Adaptation of STIM1 structure-function relationships for optogenetic control of calcium signaling. J Biol Chem 2024; 300:107636. [PMID: 39122007 PMCID: PMC11402311 DOI: 10.1016/j.jbc.2024.107636] [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: 03/12/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
In cellular contexts, the oscillation of calcium ions (Ca2+) is intricately linked to various physiological processes, such as cell proliferation, metabolism, and survival. Stromal interaction molecule 1 (STIM1) proteins form a crucial regulatory component in the store-operated calcium entry process. The structural attributes of STIM1 are vital for its functionality, encompassing distinct domains situated in the endoplasmic reticulum lumen and the cytoplasm. The intraluminal domain enables the timely detection of diminishing Ca2+ concentrations, prompting structural modifications that activate the cytoplasmic domain. This activated cytoplasmic domain undergoes conformational alterations and engages with membrane components, opening a channel that facilitates the influx of Ca2+ from the extracellular environment. Given its multiple domains and interaction mechanisms, STIM1 plays a foundational role in cellular biology. This review focuses on the design of optogenetic tools inspired by the structure and function of STIM1. These tools offer a groundbreaking approach for studying and manipulating intracellular Ca2+ signaling with precise spatiotemporal control. We further explore the practical applications of these tools, spanning fundamental scientific research, clinical studies, and their potential for translational research.
Collapse
Affiliation(s)
- Zirui Zhuang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China; School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, China
| | - Yuxin Meng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yu Xue
- School of Life Science, Tianjin University, Tianjin, China
| | - Yan Wang
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Hangzhou, China; Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ji Jing
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China; Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Maciąg F, Chhikara A, Heine M. Calcium channel signalling at neuronal endoplasmic reticulum-plasma membrane junctions. Biochem Soc Trans 2024; 52:1617-1629. [PMID: 38934485 PMCID: PMC11668288 DOI: 10.1042/bst20230819] [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: 03/27/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Neurons are highly specialised cells that need to relay information over long distances and integrate signals from thousands of synaptic inputs. The complexity of neuronal function is evident in the morphology of their plasma membrane (PM), by far the most intricate of all cell types. Yet, within the neuron lies an organelle whose architecture adds another level to this morphological sophistication - the endoplasmic reticulum (ER). Neuronal ER is abundant in the cell body and extends to distant axonal terminals and postsynaptic dendritic spines. It also adopts specialised structures like the spine apparatus in the postsynapse and the cisternal organelle in the axon initial segment. At membrane contact sites (MCSs) between the ER and the PM, the two membranes come in close proximity to create hubs of lipid exchange and Ca2+ signalling called ER-PM junctions. The development of electron and light microscopy techniques extended our knowledge on the physiological relevance of ER-PM MCSs. Equally important was the identification of ER and PM partners that interact in these junctions, most notably the STIM-ORAI and VAP-Kv2.1 pairs. The physiological functions of ER-PM junctions in neurons are being increasingly explored, but their molecular composition and the role in the dynamics of Ca2+ signalling are less clear. This review aims to outline the current state of research on the topic of neuronal ER-PM contacts. Specifically, we will summarise the involvement of different classes of Ca2+ channels in these junctions, discuss their role in neuronal development and neuropathology and propose directions for further research.
Collapse
Affiliation(s)
- Filip Maciąg
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Arun Chhikara
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| |
Collapse
|
19
|
Shi Z, Han Z, Chen J, Zhou JC. Endoplasmic reticulum-resident selenoproteins and their roles in glucose and lipid metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167246. [PMID: 38763408 DOI: 10.1016/j.bbadis.2024.167246] [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: 03/13/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Glucose and lipid metabolic disorders (GLMDs), such as diabetes, dyslipidemia, metabolic syndrome, nonalcoholic fatty liver disease, and obesity, are significant public health issues that negatively impact human health. The endoplasmic reticulum (ER) plays a crucial role at the cellular level for lipid and sterol biosynthesis, intracellular calcium storage, and protein post-translational modifications. Imbalance and dysfunction of the ER can affect glucose and lipid metabolism. As an essential trace element, selenium contributes to various human physiological functions mainly through 25 types of selenoproteins (SELENOs). At least 10 SELENOs, with experimental and/or computational evidence, are predominantly found on the ER membrane or within its lumen. Two iodothyronine deiodinases (DIOs), DIO1 and DIO2, regulate the thyroid hormone deiodination in the thyroid and some external thyroid tissues, influencing glucose and lipid metabolism. Most of the other eight members maintain redox homeostasis in the ER. Especially, SELENOF, SELENOM, and SELENOS are involved in unfolded protein responses; SELENOI catalyzes phosphatidylethanolamine synthesis; SELENOK, SELENON, and SELENOT participate in calcium homeostasis regulation; and the biological significance of thioredoxin reductase 3 in the ER remains unexplored despite its established function in the thioredoxin system. This review examines recent research advances regarding ER SELENOs in GLMDs and aims to provide insights on ER-related pathology through SELENOs regulation.
Collapse
Affiliation(s)
- Zhan Shi
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ziyu Han
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Jingyi Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Guangdong Provincial Engineering Laboratory for Nutrition Translation, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China.
| |
Collapse
|
20
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
21
|
Di Fonso A, Serano M, He M, Leigh J, Rastelli G, Dirksen RT, Protasi F, Pietrangelo L. Constitutive, Muscle-Specific Orai1 Knockout Results in the Incomplete Assembly of Ca 2+ Entry Units and a Reduction in the Age-Dependent Formation of Tubular Aggregates. Biomedicines 2024; 12:1651. [PMID: 39200116 PMCID: PMC11351919 DOI: 10.3390/biomedicines12081651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Store-operated Ca2+ entry (SOCE) is a ubiquitous cellular mechanism that cells use to activate extracellular Ca2+ entry when intracellular Ca2+ stores are depleted. In skeletal muscle, SOCE occurs within Ca2+ entry units (CEUs), intracellular junctions between stacks of SR membranes containing STIM1 and transverse tubules (TTs) containing ORAI1. Gain-of-function mutations in STIM1 and ORAI1 are linked to tubular aggregate (TA) myopathy, a disease characterized by the atypical accumulation of tubes of SR origin. Moreover, SOCE and TAs are increased in the muscles of aged male mice. Here, we assessed the longitudinal effects (from 4-6 months to 10-14 months of age) of constitutive, muscle-specific Orai1 knockout (cOrai1 KO) on skeletal muscle structure, function, and the assembly of TAs and CEUs. The results from these studies indicate that cOrai1 KO mice exhibit a shorter lifespan, reduced body weight, exercise intolerance, decreased muscle-specific force and rate of force production, and an increased number of structurally damaged mitochondria. In addition, electron microscopy analyses revealed (i) the absence of TAs with increasing age and (ii) an increased number of SR stacks without adjacent TTs (i.e., incomplete CEUs) in cOrai1 KO mice. The absence of TAs is consistent with TAs being formed as a result of excessive ORAI1-dependent Ca2+ entry.
Collapse
Affiliation(s)
- Alessia Di Fonso
- Center for Advanced Studies and Technology (CAST), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy; (A.D.F.); (M.S.); (G.R.); (F.P.)
| | - Matteo Serano
- Center for Advanced Studies and Technology (CAST), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy; (A.D.F.); (M.S.); (G.R.); (F.P.)
- Department of Medicine and Aging Sciences (DMSI), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Miao He
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (M.H.); (J.L.); (R.T.D.)
| | - Jennifer Leigh
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (M.H.); (J.L.); (R.T.D.)
| | - Giorgia Rastelli
- Center for Advanced Studies and Technology (CAST), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy; (A.D.F.); (M.S.); (G.R.); (F.P.)
- Department of Neuroscience and Clinical Sciences (DNISC), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; (M.H.); (J.L.); (R.T.D.)
| | - Feliciano Protasi
- Center for Advanced Studies and Technology (CAST), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy; (A.D.F.); (M.S.); (G.R.); (F.P.)
- Department of Medicine and Aging Sciences (DMSI), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| | - Laura Pietrangelo
- Center for Advanced Studies and Technology (CAST), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy; (A.D.F.); (M.S.); (G.R.); (F.P.)
- Department of Medicine and Aging Sciences (DMSI), University G. d’Annunzio of Chieti-Pescara, I-66100 Chieti, Italy
| |
Collapse
|
22
|
Redondo PC, Lopez JJ, Alvarado S, Jardin I, Nieto-Felipe J, Macias-Diaz A, Jimenez-Velarde V, Salido GM, Rosado JA. Extended Synaptotagmins 1 and 2 Are Required for Store-Operated Calcium Entry, Cell Migration and Viability in Breast Cancer Cells. Cancers (Basel) 2024; 16:2518. [PMID: 39061158 PMCID: PMC11274662 DOI: 10.3390/cancers16142518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Extended synaptotagmins (E-Syts) are endoplasmic reticulum (ER)-associated proteins that facilitate the tethering of the ER to the plasma membrane (PM), participating in lipid transfer between the membranes and supporting the Orai1-STIM1 interaction at ER-PM junctions. Orai1 and STIM1 are the core proteins of store-operated Ca2+ entry (SOCE), a major mechanism for Ca2+ influx that regulates a variety of cellular functions. Aberrant modulation of SOCE in cells from different types of cancer has been reported to underlie the development of several tumoral features. Here we show that estrogen receptor-positive (ER+) breast cancer MCF7 and T47D cells and triple-negative breast cancer (TNBC) MDA-MB-231 cells overexpress E-Syt1 and E-Syt2 at the protein level; the latter is also overexpressed in the TNBC BT20 cell line. E-Syt1 and E-Syt2 knockdown was without effect on SOCE in non-tumoral MCF10A breast epithelial cells and ER+ T47D breast cancer cells; however, SOCE was significantly attenuated in ER+ MCF7 cells and TNBC MDA-MB-231 and BT20 cells upon transfection with siRNA E-Syt1 or E-Syt2. Consistent with this, E-Syt1 and E-Syt2 knockdown significantly reduced cell migration and viability in ER+ MCF7 cells and the TNBC cells investigated. To summarize, E-Syt1 and E-Syt2 play a relevant functional role in breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan A. Rosado
- Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain; (J.J.L.); (S.A.); (I.J.); (J.N.-F.); (A.M.-D.); (V.J.-V.); (G.M.S.)
| |
Collapse
|
23
|
Saint-Martin Willer A, Montani D, Capuano V, Antigny F. Orai1/STIMs modulators in pulmonary vascular diseases. Cell Calcium 2024; 121:102892. [PMID: 38735127 DOI: 10.1016/j.ceca.2024.102892] [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: 02/21/2024] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
Calcium (Ca2+) is a secondary messenger that regulates various cellular processes. However, Ca2+ mishandling could lead to pathological conditions. Orai1 is a Ca2+channel contributing to the store-operated calcium entry (SOCE) and plays a critical role in Ca2+ homeostasis in several cell types. Dysregulation of Orai1 contributed to severe combined immune deficiency syndrome, some cancers, pulmonary arterial hypertension (PAH), and other cardiorespiratory diseases. During its activation process, Orai1 is mainly regulated by stromal interacting molecule (STIM) proteins, especially STIM1; however, many other regulatory partners have also been recently described. Increasing knowledge about these regulatory partners provides a better view of the downstream signalling pathways of SOCE and offers an excellent opportunity to decipher Orai1 dysregulation in these diseases. These proteins participate in other cellular functions, making them attractive therapeutic targets. This review mainly focuses on Orai1 regulatory partners in the physiological and pathological conditions of the pulmonary circulation and inflammation.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis-Robinson, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
24
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Membrane Curvature Promotes ER-PM Contact Formation via Junctophilin-EHD Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.29.601287. [PMID: 38979311 PMCID: PMC11230412 DOI: 10.1101/2024.06.29.601287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and the plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules (T-tubules), we hypothesize that the PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, while the ubiquitously expressed extended synaptotagmin 2 does not show a preference for PM curvature. At the mechanistic level, we find that the low complexity region (LCR) and the MORN motifs of junctophilins can independently bind to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins, Eps15-homology domain containing proteins (EHDs), that interact with the MORN_LCR motifs and facilitate junctophilins' preferential tethering to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a novel mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Luis A. Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Melissa L. Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Present address: Department of Nanoengineering, Jacobs School of Engineering, University of California, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Faculty of Electrical Engineering and IT, RWTH, Aachen 52074, Germany
- Institute of Biological Information Processing—Bioelectronics, IBI-3, Forschungszentrum, Juelich 52428, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| |
Collapse
|
25
|
Sallinger M, Humer C, Ong HL, Narayanasamy S, Lin QT, Fahrner M, Grabmayr H, Berlansky S, Choi S, Schmidt T, Maltan L, Atzgerstorfer L, Niederwieser M, Frischauf I, Romanin C, Stathopulos PB, Ambudkar I, Leitner R, Bonhenry D, Schindl R. Essential role of N-terminal SAM regions in STIM1 multimerization and function. Proc Natl Acad Sci U S A 2024; 121:e2318874121. [PMID: 38753510 PMCID: PMC11127010 DOI: 10.1073/pnas.2318874121] [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: 11/15/2023] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
The single-pass transmembrane protein Stromal Interaction Molecule 1 (STIM1), located in the endoplasmic reticulum (ER) membrane, possesses two main functions: It senses the ER-Ca2+ concentration and directly binds to the store-operated Ca2+ channel Orai1 for its activation when Ca2+ recedes. At high resting ER-Ca2+ concentration, the ER-luminal STIM1 domain is kept monomeric but undergoes di/multimerization once stores are depleted. Luminal STIM1 multimerization is essential to unleash the STIM C-terminal binding site for Orai1 channels. However, structural basis of the luminal association sites has so far been elusive. Here, we employed molecular dynamics (MD) simulations and identified two essential di/multimerization segments, the α7 and the adjacent region near the α9-helix in the sterile alpha motif (SAM) domain. Based on MD results, we targeted the two STIM1 SAM domains by engineering point mutations. These mutations interfered with higher-order multimerization of ER-luminal fragments in biochemical assays and puncta formation in live-cell experiments upon Ca2+ store depletion. The STIM1 multimerization impeded mutants significantly reduced Ca2+ entry via Orai1, decreasing the Ca2+ oscillation frequency as well as store-operated Ca2+ entry. Combination of the ER-luminal STIM1 multimerization mutations with gain of function mutations and coexpression of Orai1 partially ameliorated functional defects. Our data point to a hydrophobicity-driven binding within the ER-luminal STIM1 multimer that needs to switch between resting monomeric and activated multimeric state. Altogether, these data reveal that interactions between SAM domains of STIM1 monomers are critical for multimerization and activation of the protein.
Collapse
Affiliation(s)
- Matthias Sallinger
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Christina Humer
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Hwei Ling Ong
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD20892
| | - Sasirekha Narayanasamy
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD20892
| | - Qi Tong Lin
- Department of Physiology and Pharmacology, Western University, London, ONN6A5C1, Canada
| | - Marc Fahrner
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Herwig Grabmayr
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Sascha Berlansky
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Sean Choi
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD20892
| | - Tony Schmidt
- Department of Medical Physics and Biophysics, Medical University of Graz, Graz8010, Austria
| | - Lena Maltan
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Lara Atzgerstorfer
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Martin Niederwieser
- Department of Medical Physics and Biophysics, Medical University of Graz, Graz8010, Austria
| | - Irene Frischauf
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Christoph Romanin
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Peter B. Stathopulos
- Department of Physiology and Pharmacology, Western University, London, ONN6A5C1, Canada
| | - Indu Ambudkar
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD20892
| | - Romana Leitner
- Institute of Biophysics, Johannes Kepler University Linz, Linz4040, Austria
| | - Daniel Bonhenry
- Department of Physics and Materials Science, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-AlzetteL1511, Luxembourg
| | - Rainer Schindl
- Department of Medical Physics and Biophysics, Medical University of Graz, Graz8010, Austria
| |
Collapse
|
26
|
Lee JM, Kim J, Park SJ, Nam JH, Kim HJ, Kim WK. Regulation of T Lymphocyte Functions through Calcium Signaling Modulation by Nootkatone. Int J Mol Sci 2024; 25:5240. [PMID: 38791278 PMCID: PMC11121628 DOI: 10.3390/ijms25105240] [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: 04/03/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Recent advancements in understanding the intricate molecular mechanisms underlying immunological responses have underscored the critical involvement of ion channels in regulating calcium influx, particularly in inflammation. Nootkatone, a natural sesquiterpenoid found in Alpinia oxyphylla and various citrus species, has gained attention for its diverse pharmacological properties, including anti-inflammatory effects. This study aimed to elucidate the potential of nootkatone in modulating ion channels associated with calcium signaling, particularly CRAC, KV1.3, and KCa3.1 channels, which play pivotal roles in immune cell activation and proliferation. Using electrophysiological techniques, we demonstrated the inhibitory effects of nootkatone on CRAC, KV1.3, and KCa3.1 channels in HEK293T cells overexpressing respective channel proteins. Nootkatone exhibited dose-dependent inhibition of channel currents, with IC50 values determined for each channel. Nootkatone treatment did not significantly affect cell viability, indicating its potential safety for therapeutic applications. Furthermore, we observed that nootkatone treatment attenuated calcium influx through activated CRAC channels and showed anti-proliferative effects, suggesting its role in regulating inflammatory T cell activation. These findings highlight the potential of nootkatone as a natural compound for modulating calcium signaling pathways by targeting related key ion channels and it holds promise as a novel therapeutic agent for inflammatory disorders.
Collapse
Affiliation(s)
- Ji Min Lee
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Jintae Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Su Jin Park
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Hyun Jong Kim
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Gyeongsangbuk-do, Republic of Korea; (J.M.L.); (S.J.P.); (J.H.N.)
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
| | - Woo Kyung Kim
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea;
- Department of Internal Medicine, Graduate School of Medicine, Dongguk University, 27 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Gyeonggi-do, Republic of Korea
| |
Collapse
|
27
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
28
|
Fröhlich M, Söllner J, Derler I. Insights into the dynamics of the Ca2+ release-activated Ca2+ channel pore-forming complex Orai1. Biochem Soc Trans 2024; 52:747-760. [PMID: 38526208 DOI: 10.1042/bst20230815] [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: 12/20/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/26/2024]
Abstract
An important calcium (Ca2+) entry pathway into the cell is the Ca2+ release-activated Ca2+ (CRAC) channel, which controls a series of downstream signaling events such as gene transcription, secretion and proliferation. It is composed of a Ca2+ sensor in the endoplasmic reticulum (ER), the stromal interaction molecule (STIM), and the Ca2+ ion channel Orai in the plasma membrane (PM). Their activation is initiated by receptor-ligand binding at the PM, which triggers a signaling cascade within the cell that ultimately causes store depletion. The decrease in ER-luminal Ca2+ is sensed by STIM1, which undergoes structural rearrangements that lead to coupling with Orai1 and its activation. In this review, we highlight the current understanding of the Orai1 pore opening mechanism. In this context, we also point out the questions that remain unanswered and how these can be addressed by the currently emerging genetic code expansion (GCE) technology. GCE enables the incorporation of non-canonical amino acids with novel properties, such as light-sensitivity, and has the potential to provide novel insights into the structure/function relationship of CRAC channels at a single amino acid level in the living cell.
Collapse
Affiliation(s)
- Maximilian Fröhlich
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | - Julia Söllner
- 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
|
29
|
Tsomidis I, Voumvouraki A, Kouroumalis E. The Pathogenesis of Pancreatitis and the Role of Autophagy. GASTROENTEROLOGY INSIGHTS 2024; 15:303-341. [DOI: 10.3390/gastroent15020022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The pathogenesis of acute and chronic pancreatitis has recently evolved as new findings demonstrate a complex mechanism operating through various pathways. In this review, the current evidence indicating that several mechanisms act in concert to induce and perpetuate pancreatitis were presented. As autophagy is now considered a fundamental mechanism in the pathophysiology of both acute and chronic pancreatitis, the fundamentals of the autophagy pathway were discussed to allow for a better understanding of the pathophysiological mechanisms of pancreatitis. The various aspects of pathogenesis, including trypsinogen activation, ER stress and mitochondrial dysfunction, the implications of inflammation, and macrophage involvement in innate immunity, as well as the significance of pancreatic stellate cells in the development of fibrosis, were also analyzed. Recent findings on exosomes and the miRNA regulatory role were also presented. Finally, the role of autophagy in the protection and aggravation of pancreatitis and possible therapeutic implications were reviewed.
Collapse
Affiliation(s)
- Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece
| | - Elias Kouroumalis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
| |
Collapse
|
30
|
Song EAC, Chung SH, Kim JH. Molecular mechanisms of saliva secretion and hyposecretion. Eur J Oral Sci 2024; 132:e12969. [PMID: 38192116 DOI: 10.1111/eos.12969] [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: 09/11/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024]
Abstract
The exocrine salivary gland secretes saliva, a fundamental body component to maintain oral homeostasis. Saliva is composed of water, ions, and proteins such as amylase, mucins, and immunoglobulins that play essential roles in the digestion of food, lubrication, and prevention of dental caries and periodontitis. An increasing number of people experience saliva hyposecretion due to aging, medications, Sjögren's syndrome, and radiation therapy for head and neck cancer. However, current treatments are mostly limited to temporary symptomatic relief. This review explores the molecular mechanisms underlying saliva secretion and hyposecretion to provide insight into putative therapeutic targets for treatment. Proteins implicated in saliva secretion pathways, including Ca2+ -signaling proteins, aquaporins, soluble N-ethylmaleimide-sensitive factor attachment protein receptors, and tight junctions, are aberrantly expressed and localized in patients with saliva hyposecretion, such as Sjögren's syndrome. Analysis of studies on the mechanisms of saliva secretion and hyposecretion suggests that crosstalk between fluid and protein secretory pathways via Ca2+ /protein kinase C and cAMP/protein kinase A regulates saliva secretion. Impaired crosstalk between the two secretory pathways may contribute to saliva hyposecretion. Future research into the detailed regulatory mechanisms of saliva secretion and hyposecretion may provide information to define novel targets and generate therapeutic strategies for saliva hyposecretion.
Collapse
Affiliation(s)
- Eun-Ah Christine Song
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Sul-Hee Chung
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jeong Hee Kim
- Department of Oral Biochemistry and Molecular Biology, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
- Department of KHU-KIST Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Sallinger M, Grabmayr H, Humer C, Bonhenry D, Romanin C, Schindl R, Derler I. Activation mechanisms and structural dynamics of STIM proteins. J Physiol 2024; 602:1475-1507. [PMID: 36651592 DOI: 10.1113/jp283828] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The family of stromal interaction molecules (STIM) includes two widely expressed single-pass endoplasmic reticulum (ER) transmembrane proteins and additional splice variants that act as precise ER-luminal Ca2+ sensors. STIM proteins mainly function as one of the two essential components of the so-called Ca2+ release-activated Ca2+ (CRAC) channel. The second CRAC channel component is constituted by pore-forming Orai proteins in the plasma membrane. STIM and Orai physically interact with each other to enable CRAC channel opening, which is a critical prerequisite for various downstream signalling pathways such as gene transcription or proliferation. Their activation commonly requires the emptying of the intracellular ER Ca2+ store. Using their Ca2+ sensing capabilities, STIM proteins confer this Ca2+ content-dependent signal to Orai, thereby linking Ca2+ store depletion to CRAC channel opening. Here we review the conformational dynamics occurring along the entire STIM protein upon store depletion, involving the transition from the quiescent, compactly folded structure into an active, extended state, modulation by a variety of accessory components in the cell as well as the impairment of individual steps of the STIM activation cascade associated with disease.
Collapse
Affiliation(s)
- Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Herwig Grabmayr
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Christina Humer
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Daniel Bonhenry
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nove Hrady, Czech Republic
| | - Christoph Romanin
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| | - Rainer Schindl
- Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
32
|
Yao Q, Long C, Yi P, Zhang G, Wan W, Rao X, Ying J, Liang W, Hua F. C/EBPβ: A transcription factor associated with the irreversible progression of Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14721. [PMID: 38644578 PMCID: PMC11033503 DOI: 10.1111/cns.14721] [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: 11/15/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder distinguished by a swift cognitive deterioration accompanied by distinctive pathological hallmarks such as extracellular Aβ (β-amyloid) peptides, neuronal neurofibrillary tangles (NFTs), sustained neuroinflammation, and synaptic degeneration. The elevated frequency of AD cases and its proclivity to manifest at a younger age present a pressing challenge in the quest for novel therapeutic interventions. Numerous investigations have substantiated the involvement of C/EBPβ in the progression of AD pathology, thus indicating its potential as a therapeutic target for AD treatment. AIMS Several studies have demonstrated an elevation in the expression level of C/EBPβ among individuals afflicted with AD. Consequently, this review predominantly delves into the association between C/EBPβ expression and the pathological progression of Alzheimer's disease, elucidating its underlying molecular mechanism, and pointing out the possibility that C/EBPβ can be a new therapeutic target for AD. METHODS A systematic literature search was performed across multiple databases, including PubMed, Google Scholar, and so on, utilizing predetermined keywords and MeSH terms, without temporal constraints. The inclusion criteria encompassed diverse study designs, such as experimental, case-control, and cohort studies, restricted to publications in the English language, while conference abstracts and unpublished sources were excluded. RESULTS Overexpression of C/EBPβ exacerbates the pathological features of AD, primarily by promoting neuroinflammation and mediating the transcriptional regulation of key molecular pathways, including δ-secretase, apolipoprotein E4 (APOE4), acidic leucine-rich nuclear phosphoprotein-32A (ANP32A), transient receptor potential channel 1 (TRPC1), and Forkhead BoxO (FOXO). DISCUSSION The correlation between overexpression of C/EBPβ and the pathological development of AD, along with its molecular mechanisms, is evident. Investigating the pathways through which C/EBPβ regulates the development of AD reveals numerous multiple vicious cycle pathways exacerbating the pathological progression of the disease. Furthermore, the exacerbation of pathological progression due to C/EBPβ overexpression and its molecular mechanism is not limited to AD but also extends to other neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and multiple sclerosis (MS). CONCLUSION The overexpression of C/EBPβ accelerates the irreversible progression of AD pathophysiology. Additionally, C/EBPβ plays a crucial role in mediating multiple pathways linked to AD pathology, some of which engender vicious cycles, leading to the establishment of feedback mechanisms. To sum up, targeting C/EBPβ could hold promise as a therapeutic strategy not only for AD but also for other degenerative diseases.
Collapse
Affiliation(s)
- Qing Yao
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Chubing Long
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Pengcheng Yi
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Guangyong Zhang
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Wei Wan
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Xiuqin Rao
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Jun Ying
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| | - Weidong Liang
- Department of AnesthesiologyThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxi ProvinceChina
| | - Fuzhou Hua
- Department of AnesthesiologyThe Second Affiliated Hospital of Nanchang UniversityNanchang CityJiangxi ProvinceChina
- Key Laboratory of Anesthesiology of Jiangxi ProvinceNanchang CityJiangxi ProvinceChina
| |
Collapse
|
33
|
Sanchez-Lopez I, Orantos-Aguilera Y, Pozo-Guisado E, Alvarez-Barrientos A, Lilla S, Zanivan S, Lachaud C, Martin-Romero FJ. STIM1 translocation to the nucleus protects cells from DNA damage. Nucleic Acids Res 2024; 52:2389-2415. [PMID: 38224453 PMCID: PMC10954485 DOI: 10.1093/nar/gkae001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/30/2023] [Accepted: 01/01/2024] [Indexed: 01/16/2024] Open
Abstract
DNA damage represents a challenge for cells, as this damage must be eliminated to preserve cell viability and the transmission of genetic information. To reduce or eliminate unscheduled chemical modifications in genomic DNA, an extensive signaling network, known as the DNA damage response (DDR) pathway, ensures this repair. In this work, and by means of a proteomic analysis aimed at studying the STIM1 protein interactome, we have found that STIM1 is closely related to the protection from endogenous DNA damage, replicative stress, as well as to the response to interstrand crosslinks (ICLs). Here we show that STIM1 has a nuclear localization signal that mediates its translocation to the nucleus, and that this translocation and the association of STIM1 to chromatin increases in response to mitomycin-C (MMC), an ICL-inducing agent. Consequently, STIM1-deficient cell lines show higher levels of basal DNA damage, replicative stress, and increased sensitivity to MMC. We show that STIM1 normalizes FANCD2 protein levels in the nucleus, which explains the increased sensitivity of STIM1-KO cells to MMC. This study not only unveils a previously unknown nuclear function for the endoplasmic reticulum protein STIM1 but also expands our understanding of the genes involved in DNA repair.
Collapse
Affiliation(s)
- Irene Sanchez-Lopez
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Universidad de Extremadura, Badajoz 06006, Spain
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, Badajoz 06006, Spain
| | - Yolanda Orantos-Aguilera
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Universidad de Extremadura, Badajoz 06006, Spain
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, Badajoz 06006, Spain
| | - Eulalia Pozo-Guisado
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, Badajoz 06006, Spain
- Department of Cell Biology, School of Medicine, Universidad de Extremadura, Badajoz 06006, Spain
| | | | - Sergio Lilla
- CRUK Scotland Institute, Switchback Road, Glasgow G61 1BD, UK
| | - Sara Zanivan
- CRUK Scotland Institute, Switchback Road, Glasgow G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1QH, UK
| | - Christophe Lachaud
- Cancer Research Centre of Marseille, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli Calmettes, CRCM, Marseille, France
- OPALE Carnot Institute, Paris, France
| | - Francisco Javier Martin-Romero
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Universidad de Extremadura, Badajoz 06006, Spain
- Institute of Molecular Pathology Biomarkers, Universidad de Extremadura, Badajoz 06006, Spain
| |
Collapse
|
34
|
Bacsa B, Hopl V, Derler I. Synthetic Biology Meets Ca 2+ Release-Activated Ca 2+ Channel-Dependent Immunomodulation. Cells 2024; 13:468. [PMID: 38534312 PMCID: PMC10968988 DOI: 10.3390/cells13060468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Many essential biological processes are triggered by the proximity of molecules. Meanwhile, diverse approaches in synthetic biology, such as new biological parts or engineered cells, have opened up avenues to precisely control the proximity of molecules and eventually downstream signaling processes. This also applies to a main Ca2+ entry pathway into the cell, the so-called Ca2+ release-activated Ca2+ (CRAC) channel. CRAC channels are among other channels are essential in the immune response and are activated by receptor-ligand binding at the cell membrane. The latter initiates a signaling cascade within the cell, which finally triggers the coupling of the two key molecular components of the CRAC channel, namely the stromal interaction molecule, STIM, in the ER membrane and the plasma membrane Ca2+ ion channel, Orai. Ca2+ entry, established via STIM/Orai coupling, is essential for various immune cell functions, including cytokine release, proliferation, and cytotoxicity. In this review, we summarize the tools of synthetic biology that have been used so far to achieve precise control over the CRAC channel pathway and thus over downstream signaling events related to the immune response.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Division of Medical Physics und Biophysics, Medical University of Graz, A-8010 Graz, Austria;
| | - Valentina Hopl
- 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
|
35
|
Silva-Rojas R, Pérez-Guàrdia L, Simon A, Djeddi S, Treves S, Ribes A, Silva-Hernández L, Tard C, Laporte J, Böhm J. ORAI1 inhibition as an efficient preclinical therapy for tubular aggregate myopathy and Stormorken syndrome. JCI Insight 2024; 9:e174866. [PMID: 38516893 PMCID: PMC11063934 DOI: 10.1172/jci.insight.174866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) are clinically overlapping disorders characterized by childhood-onset muscle weakness and a variable occurrence of multisystemic signs, including short stature, thrombocytopenia, and hyposplenism. TAM/STRMK is caused by gain-of-function mutations in the Ca2+ sensor STIM1 or the Ca2+ channel ORAI1, both of which regulate Ca2+ homeostasis through the ubiquitous store-operated Ca2+ entry (SOCE) mechanism. Functional experiments in cells have demonstrated that the TAM/STRMK mutations induce SOCE overactivation, resulting in excessive influx of extracellular Ca2+. There is currently no treatment for TAM/STRMK, but SOCE is amenable to manipulation. Here, we crossed Stim1R304W/+ mice harboring the most common TAM/STRMK mutation with Orai1R93W/+ mice carrying an ORAI1 mutation partially obstructing Ca2+ influx. Compared with Stim1R304W/+ littermates, Stim1R304W/+Orai1R93W/+ offspring showed a normalization of bone architecture, spleen histology, and muscle morphology; an increase of thrombocytes; and improved muscle contraction and relaxation kinetics. Accordingly, comparative RNA-Seq detected more than 1,200 dysregulated genes in Stim1R304W/+ muscle and revealed a major restoration of gene expression in Stim1R304W/+Orai1R93W/+ mice. Altogether, we provide physiological, morphological, functional, and molecular data highlighting the therapeutic potential of ORAI1 inhibition to rescue the multisystemic TAM/STRMK signs, and we identified myostatin as a promising biomarker for TAM/STRMK in humans and mice.
Collapse
Affiliation(s)
- Roberto Silva-Rojas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Laura Pérez-Guàrdia
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Alix Simon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Sarah Djeddi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Susan Treves
- Departments of Neurology and Biomedicine, Basel University Hospital, Basel, Switzerland
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Agnès Ribes
- Institute of Metabolic and Cardiovascular Disease, Inserm UMR1297 and University of Toulouse 3, Toulouse, France
- Laboratory of Hematology, University Hospital of Toulouse, Toulouse, France
| | - Lorenzo Silva-Hernández
- Neurology Service, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Céline Tard
- University Lille, Inserm, CHU Lille, U1172 Lille Neuroscience & Cognition, Center for Rare Neuromuscular Diseases Nord/Est/Ile-de-France, Lille, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, University of Strasbourg, Illkirch, France
| |
Collapse
|
36
|
Ji R, Chang L, An C, Zhang J. Proton-sensing ion channels, GPCRs and calcium signaling regulated by them: implications for cancer. Front Cell Dev Biol 2024; 12:1326231. [PMID: 38505262 PMCID: PMC10949864 DOI: 10.3389/fcell.2024.1326231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Extracellular acidification of tumors is common. Through proton-sensing ion channels or proton-sensing G protein-coupled receptors (GPCRs), tumor cells sense extracellular acidification to stimulate a variety of intracellular signaling pathways including the calcium signaling, which consequently exerts global impacts on tumor cells. Proton-sensing ion channels, and proton-sensing GPCRs have natural advantages as drug targets of anticancer therapy. However, they and the calcium signaling regulated by them attracted limited attention as potential targets of anticancer drugs. In the present review, we discuss the progress in studies on proton-sensing ion channels, and proton-sensing GPCRs, especially emphasizing the effects of calcium signaling activated by them on the characteristics of tumors, including proliferation, migration, invasion, metastasis, drug resistance, angiogenesis. In addition, we review the drugs targeting proton-sensing channels or GPCRs that are currently in clinical trials, as well as the relevant potential drugs for cancer treatments, and discuss their future prospects. The present review aims to elucidate the important role of proton-sensing ion channels, GPCRs and calcium signaling regulated by them in cancer initiation and development. This review will promote the development of drugs targeting proton-sensing channels or GPCRs for cancer treatments, effectively taking their unique advantage as anti-cancer drug targets.
Collapse
Affiliation(s)
- Renhui Ji
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Li Chang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
- Department of Pathophysiology, Basic Medicine College of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| | - Junjing Zhang
- Foundational and Translational Medical Research Center, Department of Allergy and General Surgery, Hohhot First Hospital, Hohhot, China
| |
Collapse
|
37
|
Michalak M. Calreticulin: Endoplasmic reticulum Ca 2+ gatekeeper. J Cell Mol Med 2024; 28:e17839. [PMID: 37424156 PMCID: PMC10902585 DOI: 10.1111/jcmm.17839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Endoplasmic reticulum (ER) luminal Ca2+ is vital for the function of the ER and regulates many cellular processes. Calreticulin is a highly conserved, ER-resident Ca2+ binding protein and lectin-like chaperone. Over four decades of studying calreticulin demonstrate that this protein plays a crucial role in maintaining Ca2+ supply under different physiological conditions, in managing access to Ca2+ and how Ca2+ is used depending on the environmental events and in making sure that Ca2+ is not misused. Calreticulin plays a role of ER luminal Ca2+ sensor to manage Ca2+-dependent ER luminal events including maintaining interaction with its partners, Ca2+ handling molecules, substrates and stress sensors. The protein is strategically positioned in the lumen of the ER from where the protein manages access to and distribution of Ca2+ for many cellular Ca2+-signalling events. The importance of calreticulin Ca2+ pool extends beyond the ER and includes influence of cellular processes involved in many aspects of cellular pathophysiology. Abnormal handling of the ER Ca2+ contributes to many pathologies from heart failure to neurodegeneration and metabolic diseases.
Collapse
Affiliation(s)
- Marek Michalak
- Department of BiochemistryUniversity of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
38
|
Song SE, Shin SK, Ju HY, Im SS, Song DK. Role of cytosolic and endoplasmic reticulum Ca 2+ in pancreatic beta-cells: pros and cons. Pflugers Arch 2024; 476:151-161. [PMID: 37940681 DOI: 10.1007/s00424-023-02872-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
Pancreatic beta cells utilize Ca2+ to secrete insulin in response to glucose. The glucose-dependent increase in cytosolic Ca2+ concentration ([Ca2+]C) activates a series of insulin secretory machinery in pancreatic beta cells. Therefore, the amount of insulin secreted in response to glucose is determined in a [Ca2+]C-dependent manner, at least within a moderate range. However, the demand for insulin secretion may surpass the capability of beta cells. Abnormal elevation of [Ca2+]C levels beyond the beta-cell endurance capacity can damage them by inducing endoplasmic reticulum (ER) stress and cell death programs such as apoptosis. Therefore, while Ca2+ is essential for the insulin secretory functions of beta cells, it could affect their survival at pathologically higher levels. Because an increase in beta-cell [Ca2+]C is inevitable under certain hazardous conditions, understanding the regulatory mechanism for [Ca2+]C is important. Therefore, this review discusses beta-cell function, survival, ER stress, and apoptosis associated with intracellular and ER Ca2+ homeostasis.
Collapse
Affiliation(s)
- Seung-Eun Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, South Korea
| | - Hyeon Yeong Ju
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung-Soon Im
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Dae-Kyu Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea.
| |
Collapse
|
39
|
Sun S, Zhao G, Jia M, Jiang Q, Li S, Wang H, Li W, Wang Y, Bian X, Zhao YG, Huang X, Yang G, Cai H, Pastor-Pareja JC, Ge L, Zhang C, Hu J. Stay in touch with the endoplasmic reticulum. SCIENCE CHINA. LIFE SCIENCES 2024; 67:230-257. [PMID: 38212460 DOI: 10.1007/s11427-023-2443-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/28/2023] [Indexed: 01/13/2024]
Abstract
The endoplasmic reticulum (ER), which is composed of a continuous network of tubules and sheets, forms the most widely distributed membrane system in eukaryotic cells. As a result, it engages a variety of organelles by establishing membrane contact sites (MCSs). These contacts regulate organelle positioning and remodeling, including fusion and fission, facilitate precise lipid exchange, and couple vital signaling events. Here, we systematically review recent advances and converging themes on ER-involved organellar contact. The molecular basis, cellular influence, and potential physiological functions for ER/nuclear envelope contacts with mitochondria, Golgi, endosomes, lysosomes, lipid droplets, autophagosomes, and plasma membrane are summarized.
Collapse
Affiliation(s)
- Sha Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gan Zhao
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Mingkang Jia
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haibin Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenjing Li
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunyun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xin Bian
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yan G Zhao
- Brain Research Center, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ge Yang
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jose C Pastor-Pareja
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Neurosciences, Consejo Superior de Investigaciones Cientfflcas-Universidad Miguel Hernandez, San Juan de Alicante, 03550, Spain.
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
40
|
Zhang H, Feng Y, Si Y, Lu C, Wang J, Wang S, Li L, Xie W, Yue Z, Yong J, Dai S, Zhang L, Li X. Shank3 ameliorates neuronal injury after cerebral ischemia/reperfusion via inhibiting oxidative stress and inflammation. Redox Biol 2024; 69:102983. [PMID: 38064762 PMCID: PMC10755590 DOI: 10.1016/j.redox.2023.102983] [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: 10/16/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 01/01/2024] Open
Abstract
Shank3, a key molecule related to the development and deterioration of autism, has recently been found to downregulate in the murine brain after ischemia/reperfusion (I/R). Despite this discovery, however, its effects on neuronal injury and the mechanism underlying the effects remain to be clarified. To address this, in this study, based on genetically modified mice models, we revealed that the expression of Shank3 showed a time-dependent change in murine hippocampal neurons after I/R, and that conditional knockout (cko) of Shank3 in neurons resulted in aggravated neuronal injuries. The protective effects of Shank3 against oxidative stress and inflammation after I/R were achieved through direct binding STIM1 and subsequent proteasome-mediated degradation of STIM1. The STIM1 downregulation induced the phosphorylation of downstream Nrf2 Ser40, which subsequently translocated to the nucleus, and further increased the expression of antioxidant genes such as NQO1 and HO-1 in HT22 cells. In vivo, the study has further confirmed that double knockout of Shank3 and Stim1 alleviated oxidative stress and inflammation after I/R in Shank3cko mice. In conclusion, the present study has demonstrated that Shank3 interacts with STIM1 and inhibits post-I/R neuronal oxidative stress and inflammatory response via the Nrf2 pathway. This interaction can potentially contribute to the development of a promising method for I/R treatment.
Collapse
Affiliation(s)
- Hongchen Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Feng
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanfang Si
- Department of Ophthalmology, The Eighth Medical Center, Affiliated to the Senior Department of Ophthalmology, The Third Medical Center, Chinese People's Liberation Army General Hospital, Beijing, 100091, China
| | - Chuanhao Lu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiquan Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Liang Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenyu Xie
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheming Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Yong
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
41
|
Bryson V, Wang C, Zhou Z, Singh K, Volin N, Yildirim E, Rosenberg P. The D84G mutation in STIM1 causes nuclear envelope dysfunction and myopathy in mice. J Clin Invest 2024; 134:e170317. [PMID: 38300705 PMCID: PMC10977986 DOI: 10.1172/jci170317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 01/26/2024] [Indexed: 02/03/2024] Open
Abstract
Stromal interaction molecule 1 (STIM1) is a Ca2+ sensor located in the sarcoplasmic reticulum (SR) of skeletal muscle, where it is best known for its role in store-operated Ca2+ entry (SOCE). Genetic syndromes resulting from STIM1 mutations are recognized as a cause of muscle weakness and atrophy. Here, we focused on a gain-of-function mutation that occurs in humans and mice (STIM1+/D84G mice), in which muscles exhibited constitutive SOCE. Unexpectedly, this constitutive SOCE did not affect global Ca2+ transients, SR Ca2+ content, or excitation-contraction coupling (ECC) and was therefore unlikely to underlie the reduced muscle mass and weakness observed in these mice. Instead, we demonstrate that the presence of D84G STIM1 in the nuclear envelope of STIM1+/D84G muscle disrupted nuclear-cytosolic coupling, causing severe derangement in nuclear architecture, DNA damage, and altered lamina A-associated gene expression. Functionally, we found that D84G STIM1 reduced the transfer of Ca2+ from the cytosol to the nucleus in myoblasts, resulting in a reduction of [Ca2+]N. Taken together, we propose a novel role for STIM1 in the nuclear envelope that links Ca2+ signaling to nuclear stability in skeletal muscle.
Collapse
Affiliation(s)
| | - Chaojian Wang
- Department of Medicine
- Duke Cardiovascular Research Center
| | | | | | | | - Eda Yildirim
- Department of Cell Biology
- Duke Cancer Institute, Duke University Medical Center, and
| | - Paul Rosenberg
- Department of Medicine
- Duke Cardiovascular Research Center
- Duke Molecular Physiology Institute, School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
42
|
Voeltz GK, Sawyer EM, Hajnóczky G, Prinz WA. Making the connection: How membrane contact sites have changed our view of organelle biology. Cell 2024; 187:257-270. [PMID: 38242082 DOI: 10.1016/j.cell.2023.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 01/21/2024]
Abstract
The view of organelles and how they operate together has changed dramatically over the last two decades. The textbook view of organelles was that they operated largely independently and were connected by vesicular trafficking and the diffusion of signals through the cytoplasm. We now know that all organelles make functional close contacts with one another, often called membrane contact sites. The study of these sites has moved to center stage in cell biology as it has become clear that they play critical roles in healthy and developing cells and during cell stress and disease states. Contact sites have important roles in intracellular signaling, lipid metabolism, motor-protein-mediated membrane dynamics, organelle division, and organelle biogenesis. Here, we summarize the major conceptual changes that have occurred in cell biology as we have come to appreciate how contact sites integrate the activities of organelles.
Collapse
Affiliation(s)
- G K Voeltz
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - E M Sawyer
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - G Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - W A Prinz
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
43
|
Khanna K, Yan H, Mehra M, Rohatgi N, Mbalaviele G, Mellins ED, Faccio R. Tmem178 Negatively Regulates IL-1β Production Through Inhibition of the NLRP3 Inflammasome. Arthritis Rheumatol 2024; 76:107-118. [PMID: 37534578 PMCID: PMC11421209 DOI: 10.1002/art.42666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/30/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE Inflammasomes modulate the release of bioactive interleukin (IL)-1β. Excessive IL-1β levels are detected in patients with systemic juvenile idiopathic arthritis (sJIA) and cytokine storm syndrome (CSS) with mutated and unmutated inflammasome components, raising questions on the mechanisms of IL-1β regulation in these disorders. METHODS To investigate how the NLRP3 inflammasome is modulated in sJIA, we focused on Transmembrane protein 178 (Tmem178), a negative regulator of calcium levels in macrophages, and measured IL-1β and caspase-1 activation in wild-type (WT) and Tmem178-/- macrophages after calcium chelators, silencing of Stim1, a component of store-operated calcium entry (SOCE), or by expressing a Tmem178 mutant lacking the Stromal Interaction Molecule 1 (Stim1) binding site. Mitochondrial function in both genotypes was assessed by measuring oxidative respiration, mitochondrial reactive oxygen species (mtROS), and mitochondrial damage. CSS development was analyzed in Perforin-/- /Tmem178-/- mice infected with lymphocytic choriomeningitis virus (LCMV) in which inflammasome or IL-1β signaling was pharmacologically inhibited. Human TMEM178 and IL1B transcripts were analyzed in data sets of whole blood and peripheral blood monocytes from healthy controls and patients with active sJIA. RESULTS TMEM178 levels are reduced in whole blood and monocytes from patients with sJIA while IL1B levels are increased. Accordingly, Tmem178-/- macrophages produce elevated IL-1β compared with WT cells. The elevated intracellular calcium levels after SOCE activation in Tmem178-/- macrophages induce mitochondrial damage, release mtROS, and ultimately promote NLRP3 inflammasome activation. In vivo, inhibition of inflammasome or IL-1β neutralization prolongs Tmem178-/- mouse survival in LCMV-induced CSS. CONCLUSION Down-regulation of TMEM178 levels may represent a marker of disease activity and help identify patients who could benefit from inflammasome targeting.
Collapse
Affiliation(s)
- Kunjan Khanna
- Department of Orthopedics, Washington University in St Louis, MO, USA
- These authors contributed equally
| | - Hui Yan
- Department of Orthopedics, Washington University in St Louis, MO, USA
- Current address: Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- These authors contributed equally
| | - Muneshwar Mehra
- Department of Neuroscience, Washington University in St Louis, MO, USA
| | - Nidhi Rohatgi
- Department of Pathology and Immunology, Washington University in St Louis, MO, USA
| | | | | | - Roberta Faccio
- Department of Orthopedics, Washington University in St Louis, MO, USA
- Shriners Hospital for Children, St Louis, MO, USA
| |
Collapse
|
44
|
Kohler A, Kohler V. Better Together: Interorganellar Communication in the Regulation of Proteostasis. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241272245. [PMID: 39385949 PMCID: PMC11462569 DOI: 10.1177/25152564241272245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 10/12/2024]
Abstract
An extensive network of chaperones and folding factors is responsible for maintaining a functional proteome, which is the basis for cellular life. The underlying proteostatic mechanisms are not isolated within organelles, rather they are connected over organellar borders via signalling processes or direct association via contact sites. This review aims to provide a conceptual understanding of proteostatic mechanisms across organelle borders, not focussing on individual organelles. This discussion highlights the precision of these finely tuned systems, emphasising the complicated balance between cellular protection and adaptation to stress. In this review, we discuss widely accepted aspects while shedding light on newly discovered perspectives.
Collapse
Affiliation(s)
- Andreas Kohler
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87 Umeå, Sweden
| | - Verena Kohler
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
45
|
Kong X, Wang F, Chen Y, Liang X, Yin Y, Liu H, Luo G, Li Y, Liang S, Wang Y, Liu Z, Tang C. Molecular action mechanisms of two novel and selective calcium release-activated calcium channel antagonists. Int J Biol Macromol 2023; 253:126937. [PMID: 37722647 DOI: 10.1016/j.ijbiomac.2023.126937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
The prototypical calcium release-activated calcium (CRAC) channel, composed of STIM1 and Orai1, is a sought-after drug target for treating autoimmune disorders. Herein, we identified two novel and selective CRAC channel inhibitors, the indole-like compound C63368 and pyrazole core-containing compound C79413, potently and reversibly inhibiting the CRAC channel with low micromolar IC50s and sparing various off-target ion channels. These two compounds did not inhibit STIM1 activation or its coupling with Orai1, nor did they affect the channel's calcium-dependent fast inactivation. Instead, they directly acted on the Orai1 protein, with the channel's pore geometry profoundly affecting their potencies. In vitro, C63368 and C79413 effectively inhibited Jurkat cell proliferation and cytokines production in human T lymphocytes. Intragastric administration of C63368 and C79413 to mice yielded great therapeutic benefits in psoriasis and colitis animal models of autoimmune disorders, reducing serum cytokines production and significantly relieving pathological symptoms. It's worth noting, that this study provided the first insight into the characterization and mechanistic investigation of an indole-like CRAC channel antagonist. Altogether, the identification of these two highly selective CRAC channel antagonists, coupled with the elucidation of their action mechanisms, not only provides valuable template molecules but also offers profound insights for drug development targeting the CRAC channel.
Collapse
Affiliation(s)
- Xiangjin Kong
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China
| | - Feifan Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Yan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Xinyao Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuan Yin
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Hao Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Guoqing Luo
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yinping Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Songping Liang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China.
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Changsha 40081, China.
| |
Collapse
|
46
|
Carreras-Sureda A, Zhang X, Laubry L, Brunetti J, Koenig S, Wang X, Castelbou C, Hetz C, Liu Y, Frieden M, Demaurex N. The ER stress sensor IRE1 interacts with STIM1 to promote store-operated calcium entry, T cell activation, and muscular differentiation. Cell Rep 2023; 42:113540. [PMID: 38060449 DOI: 10.1016/j.celrep.2023.113540] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/29/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE) mediated by stromal interacting molecule (STIM)-gated ORAI channels at endoplasmic reticulum (ER) and plasma membrane (PM) contact sites maintains adequate levels of Ca2+ within the ER lumen during Ca2+ signaling. Disruption of ER Ca2+ homeostasis activates the unfolded protein response (UPR) to restore proteostasis. Here, we report that the UPR transducer inositol-requiring enzyme 1 (IRE1) interacts with STIM1, promotes ER-PM contact sites, and enhances SOCE. IRE1 deficiency reduces T cell activation and human myoblast differentiation. In turn, STIM1 deficiency reduces IRE1 signaling after store depletion. Using a CaMPARI2-based Ca2+ genome-wide screen, we identify CAMKG2 and slc105a as SOCE enhancers during ER stress. Our findings unveil a direct crosstalk between SOCE and UPR via IRE1, acting as key regulator of ER Ca2+ and proteostasis in T cells and muscles. Under ER stress, this IRE1-STIM1 axis boosts SOCE to preserve immune cell functions, a pathway that could be targeted for cancer immunotherapy.
Collapse
Affiliation(s)
- Amado Carreras-Sureda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| | - Xin Zhang
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Loann Laubry
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Jessica Brunetti
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Stéphane Koenig
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Xiaoxia Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cyril Castelbou
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience (GERO), Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Maud Frieden
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
47
|
He Y, Liu T, He Q, Ke W, Li X, Du J, Deng S, Shu Z, Wu J, Yang B, Wang Y, Mao Y, Rao Y, Shu Y, Peng B. Microglia facilitate and stabilize the response to general anesthesia via modulating the neuronal network in a brain region-specific manner. eLife 2023; 12:RP92252. [PMID: 38131301 PMCID: PMC10746144 DOI: 10.7554/elife.92252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
General anesthesia leads to a loss of consciousness and an unrousable state in patients. Although general anesthetics are widely used in clinical practice, their underlying mechanisms remain elusive. The potential involvement of nonneuronal cells is unknown. Microglia are important immune cells in the central nervous system (CNS) that play critical roles in CNS function and dysfunction. We unintentionally observed delayed anesthesia induction and early anesthesia emergence in microglia-depleted mice. We found that microglial depletion differentially regulates neuronal activities by suppressing the neuronal network of anesthesia-activated brain regions and activating emergence-activated brain regions. Thus, microglia facilitate and stabilize the anesthesia status. This influence is not mediated by dendritic spine plasticity. Instead, it relies on the activation of microglial P2Y12 and subsequent calcium influx, which facilitates the general anesthesia response. Together, we elucidate the regulatory role of microglia in general anesthesia, extending our knowledge of how nonneuronal cells modulate neuronal activities.
Collapse
Affiliation(s)
- Yang He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Taohui Liu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Wei Ke
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Xiaoyu Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jinjin Du
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Suixin Deng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Zhenfeng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Jialin Wu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Baozhi Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Yuqing Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- School of Basic Medical Sciences, Jinzhou Medical UniversityJinzhouChina
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Yanxia Rao
- Department of Neurology, Zhongshan Hospital, Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan UniversityShanghaiChina
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan UniversityShanghaiChina
- Co-Innovation Center of Neurodegeneration, Nantong UniversityNantongChina
| |
Collapse
|
48
|
Ramlow L, Falcke M, Lindner B. An integrate-and-fire approach to Ca 2+ signaling. Part II: Cumulative refractoriness. Biophys J 2023; 122:4710-4729. [PMID: 37981761 PMCID: PMC10754692 DOI: 10.1016/j.bpj.2023.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/20/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023] Open
Abstract
Inositol 1,4,5-trisphosphate-induced Ca2+ signaling is a second messenger system used by almost all eukaryotic cells. The agonist concentration stimulating Ca2+ signals is encoded in the frequency of a Ca2+ concentration spike sequence. When a cell is stimulated, the interspike intervals (ISIs) often show a distinct transient during which they gradually increase, a system property we refer to as cumulative refractoriness. We extend a previously published stochastic model to include the Ca2+ concentration in the intracellular Ca2+ store as a slow adaptation variable. This model can reproduce both stationary and transient statistics of experimentally observed ISI sequences. We derive approximate expressions for the mean and coefficient of variation of the stationary ISIs. We also consider the response to the onset of a constant stimulus and estimate the length of the transient and the strength of the adaptation of the ISI. We show that the adaptation sets the coefficient of variation in agreement with current ideas derived from experiments. Moreover, we explain why, despite a pronounced transient behavior, ISI correlations can be weak, as often observed in experiments. Finally, we fit our model to reproduce the transient statistics of experimentally observed ISI sequences in stimulated HEK cells. The fitted model is able to qualitatively reproduce the relationship between the stationary interval correlations and the number of transient intervals, as well as the strength of the ISI adaptation. We also find positive correlations in the experimental sequence that cannot be explained by our model.
Collapse
Affiliation(s)
- Lukas Ramlow
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany; Department of Physics, Humboldt University Berlin, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Martin Falcke
- Department of Physics, Humboldt University Berlin, Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| | - Benjamin Lindner
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany; Department of Physics, Humboldt University Berlin, Berlin, Germany
| |
Collapse
|
49
|
Kodakandla G, Akimzhanov AM, Boehning D. Regulatory mechanisms controlling store-operated calcium entry. Front Physiol 2023; 14:1330259. [PMID: 38169682 PMCID: PMC10758431 DOI: 10.3389/fphys.2023.1330259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Calcium influx through plasma membrane ion channels is crucial for many events in cellular physiology. Cell surface stimuli lead to the production of inositol 1,4,5-trisphosphate (IP3), which binds to IP3 receptors (IP3R) in the endoplasmic reticulum (ER) to release calcium pools from the ER lumen. This leads to the depletion of ER calcium pools, which has been termed store depletion. Store depletion leads to the dissociation of calcium ions from the EF-hand motif of the ER calcium sensor Stromal Interaction Molecule 1 (STIM1). This leads to a conformational change in STIM1, which helps it to interact with the plasma membrane (PM) at ER:PM junctions. At these ER:PM junctions, STIM1 binds to and activates a calcium channel known as Orai1 to form calcium release-activated calcium (CRAC) channels. Activation of Orai1 leads to calcium influx, known as store-operated calcium entry (SOCE). In addition to Orai1 and STIM1, the homologs of Orai1 and STIM1, such as Orai2/3 and STIM2, also play a crucial role in calcium homeostasis. The influx of calcium through the Orai channel activates a calcium current that has been termed the CRAC current. CRAC channels form multimers and cluster together in large macromolecular assemblies termed "puncta". How CRAC channels form puncta has been contentious since their discovery. In this review, we will outline the history of SOCE, the molecular players involved in this process, as well as the models that have been proposed to explain this critical mechanism in cellular physiology.
Collapse
Affiliation(s)
- Goutham Kodakandla
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| | - Askar M. Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, Houston, TX, United States
| | - Darren Boehning
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, United States
| |
Collapse
|
50
|
Kodakandla G, Akimzhanov AM, Boehning D. Regulatory mechanisms controlling store-operated calcium entry. ARXIV 2023:arXiv:2309.06907v3. [PMID: 37744466 PMCID: PMC10516112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Calcium influx through plasma membrane ion channels is crucial for many events in cellular physiology. Cell surface stimuli lead to the production of inositol 1,4,5-trisphosphate (IP3), which binds to IP3 receptors (IP3R) in the endoplasmic reticulum (ER) to release calcium pools from the ER lumen. This leads to the depletion of ER calcium pools, which has been termed store depletion. Store depletion leads to the dissociation of calcium ions from the EF-hand motif of the ER calcium sensor Stromal Interaction Molecule 1 (STIM1). This leads to a conformational change in STIM1, which helps it to interact with the plasma membrane (PM) at ER:PM junctions. At these ER:PM junctions, STIM1 binds to and activates a calcium channel known as Orai1 to form calcium-release activated calcium (CRAC) channels. Activation of Orai1 leads to calcium influx, known as store-operated calcium entry (SOCE). In addition to Orai1 and STIM1, the homologs of Orai1 and STIM1, such as Orai2/3 and STIM2, also play a crucial role in calcium homeostasis. The influx of calcium through the Orai channel activates a calcium current that has been termed the CRAC current. CRAC channels form multimers and cluster together in large macromolecular assemblies termed "puncta". How CRAC channels form puncta has been contentious since their discovery. In this review, we will outline the history of SOCE, the molecular players involved in this process, as well as the models that have been proposed to explain this critical mechanism in cellular physiology.
Collapse
Affiliation(s)
- Goutham Kodakandla
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA, 08103
| | - Askar M. Akimzhanov
- Department of Biochemistry and Molecular Biology, McGovern Medical School, Houston, Texas, USA, 77030
| | - Darren Boehning
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA, 08103
| |
Collapse
|