51
|
Wani K, AlHarthi H, Alghamdi A, Sabico S, Al-Daghri NM. Role of NLRP3 Inflammasome Activation in Obesity-Mediated Metabolic Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:E511. [PMID: 33435142 PMCID: PMC7826517 DOI: 10.3390/ijerph18020511] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
NLRP3 inflammasome is one of the multimeric protein complexes of the nucleotide-binding domain, leucine-rich repeat (NLR)-containing pyrin and HIN domain family (PYHIN). When activated, NLRP3 inflammasome triggers the release of pro-inflammatory interleukins (IL)-1β and IL-18, an essential step in innate immune response; however, defective checkpoints in inflammasome activation may lead to autoimmune, autoinflammatory, and metabolic disorders. Among the consequences of NLRP3 inflammasome activation is systemic chronic low-grade inflammation, a cardinal feature of obesity and insulin resistance. Understanding the mechanisms involved in the regulation of NLRP3 inflammasome in adipose tissue may help in the development of specific inhibitors for the treatment and prevention of obesity-mediated metabolic diseases. In this narrative review, the current understanding of NLRP3 inflammasome activation and regulation is highlighted, including its putative roles in adipose tissue dysfunction and insulin resistance. Specific inhibitors of NLRP3 inflammasome activation which can potentially be used to treat metabolic disorders are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Nasser M. Al-Daghri
- Biochemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (K.W.); (H.A.); (A.A.); (S.S.)
| |
Collapse
|
52
|
Pagliaro L, Marchesini M, Roti G. Targeting oncogenic Notch signaling with SERCA inhibitors. J Hematol Oncol 2021; 14:8. [PMID: 33407740 PMCID: PMC7789735 DOI: 10.1186/s13045-020-01015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.
Collapse
Affiliation(s)
- Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Matteo Marchesini
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy.
| |
Collapse
|
53
|
Ismatullah H, Jabeen I, Saeed MT. Biological Regulatory Network (BRN) Analysis and Molecular Docking Simulations to Probe the Modulation of IP 3R Mediated Ca 2+ Signaling in Cancer. Genes (Basel) 2020; 12:34. [PMID: 33383780 PMCID: PMC7823498 DOI: 10.3390/genes12010034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
Inositol trisphosphate receptor (IP3R) mediated Ca+2 signaling is essential in determining the cell fate by regulating numerous cellular processes, including cell division and cell death. Despite extensive studies about the characterization of IP3R in cancer, the underlying molecular mechanism initiating the cell proliferation and apoptosis remained enigmatic. Moreover, in cancer, the modulation of IP3R in downstream signaling pathways, which control oncogenesis and cancer progression, is not well characterized. Here, we constructed a biological regulatory network (BRN), and describe the remodeling of IP3R mediated Ca2+ signaling as a central key that controls the cellular processes in cancer. Moreover, we summarize how the inhibition of IP3R affects the deregulated cell proliferation and cell death in cancer cells and results in the initiation of pro-survival responses in resistance of cell death in normal cells. Further, we also investigated the role of stereo-specificity of IP3 molecule and its analogs in binding with the IP3 receptor. Molecular docking simulations showed that the hydroxyl group at R6 position along with the phosphate group at R5 position in 'R' conformation is more favorable for IP3 interactions. Additionally, Arg-266 and Arg-510 showed π-π and hydrogen bond interactions and Ser-278 forms hydrogen bond interactions with the IP3 binding site. Thus, they are identified as crucial for the binding of antagonists.
Collapse
Affiliation(s)
| | - Ishrat Jabeen
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), Academic-I Building, H-12 Islamabad 44000, Pakistan; (H.I.); (M.T.S.)
| | | |
Collapse
|
54
|
Chiarelli N, Zoppi N, Ritelli M, Venturini M, Capitanio D, Gelfi C, Colombi M. Biological insights in the pathogenesis of hypermobile Ehlers-Danlos syndrome from proteome profiling of patients' dermal myofibroblasts. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166051. [PMID: 33383104 DOI: 10.1016/j.bbadis.2020.166051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
Hypermobile Ehlers-Danlos syndrome (hEDS), mainly characterized by generalized joint hypermobility and its complications, minor skin changes, and apparently segregating with an autosomal dominant pattern, is still without a known molecular basis. Hence, its diagnosis is only clinical based on a strict set of criteria defined in the revised EDS nosology. Moreover, the hEDS phenotypic spectrum is wide-ranging and comprises multiple associated signs and symptoms shared with other heritable or acquired connective tissue disorders and chronic inflammatory diseases. In this complex scenario, we previously demonstrated that hEDS patients' skin fibroblasts show phenotypic features of myofibroblasts, widespread extracellular matrix (ECM) disarray, perturbation of ECM-cell contacts, and dysregulated expression of genes involved in connective tissue architecture and related to inflammatory and pain responses. Herein, the cellular proteome of 6 hEDS dermal myofibroblasts was compared to that of 12 control fibroblasts to deepen the knowledge on mechanisms involved in the disease pathogenesis. Qualitative and quantitative differences were assessed based on top-down and bottom-up approaches and some differentially expressed proteins were proofed by biochemical analyses. Proteomics disclosed the differential expression of proteins principally implicated in cytoskeleton organization, energy metabolism and redox balance, proteostasis, and intracellular trafficking. Our findings offer a comprehensive view of dysregulated protein networks and related pathways likely associated with the hEDS pathophysiology. The present results can be regarded as a starting point for future in-depth investigations aimed to decipher the functional impact of potential bioactive molecules for the development of targeted management and therapies.
Collapse
Affiliation(s)
- Nicola Chiarelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Venturini
- Division of Dermatology, Department of Clinical and Experimental Sciences, Spedali Civili University Hospital Brescia, Italy
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy; IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
55
|
Various Aspects of Calcium Signaling in the Regulation of Apoptosis, Autophagy, Cell Proliferation, and Cancer. Int J Mol Sci 2020; 21:ijms21218323. [PMID: 33171939 PMCID: PMC7664196 DOI: 10.3390/ijms21218323] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca2+) is a major second messenger in cells and is essential for the fate and survival of all higher organisms. Different Ca2+ channels, pumps, or exchangers regulate variations in the duration and levels of intracellular Ca2+, which may be transient or sustained. These changes are then decoded by an elaborate toolkit of Ca2+-sensors, which translate Ca2+ signal to intracellular operational cell machinery, thereby regulating numerous Ca2+-dependent physiological processes. Alterations to Ca2+ homoeostasis and signaling are often deleterious and are associated with certain pathological states, including cancer. Altered Ca2+ transmission has been implicated in a variety of processes fundamental for the uncontrolled proliferation and invasiveness of tumor cells and other processes important for cancer progression, such as the development of resistance to cancer therapies. Here, we review what is known about Ca2+ signaling and how this fundamental second messenger regulates life and death decisions in the context of cancer, with particular attention directed to cell proliferation, apoptosis, and autophagy. We also explore the intersections of Ca2+ and the therapeutic targeting of cancer cells, summarizing the therapeutic opportunities for Ca2+ signal modulators to improve the effectiveness of current anticancer therapies.
Collapse
|
56
|
De Loof A, Schoofs L. Two Undervalued Functions of the Golgi Apparatus: Removal of Excess Ca 2+ and Biosynthesis of Farnesol-Like Sesquiterpenoids, Possibly as Ca 2+-Pump Agonists and Membrane "Fluidizers-Plasticizers". Front Physiol 2020; 11:542879. [PMID: 33178030 PMCID: PMC7593688 DOI: 10.3389/fphys.2020.542879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
The extensive literature dealing with the Golgi system emphasizes its role in protein secretion and modification, usually without specifying from which evolutionary ancient cell physiological necessity such secretion originated. Neither does it specify which functional requirements the secreted proteins must meet. From a reinterpretation of some classical and recent data gained mainly, but not exclusively, from (insect) endocrinology, the view emerged that the likely primordial function of the rough endoplasmic reticulum (RER)–Golgi complex in all eukaryotes was not the secretion of any type of protein but the removal of toxic excess Ca2+ from the cytoplasm. Such activity requires the concurrent secretion of large amounts of Ca2+-carrying/transporting proteins acting as a micro-conveyor belt system inside the RER–Golgi. Thus, (fitness increasing) protein secretion is subordinate to Ca2+ removal. Milk with its high content of protein and Ca2+ (60–90 mM vs. 100 nM in unstimulated mammary gland cells) is an extreme example. The sarco(endo)plasmatic reticulum Ca2+-ATPases (SERCAs) and SPCA1a Ca2+/Mn2+ transport ATPases are major players in Ca2+ removal through the Golgi. Both are blocked by the sesquiterpenoid thapsigargin. This strengthens the hypothesis (2014) that endogenous farnesol-like sesquiterpenoids (FLSs) may act as the long sought for but still unidentified agonist(s) for Ca2+-pumps in both the ER and Golgi. A second putative function also emerges. The fusion of both the incoming and outgoing transport vesicles, respectively, at the cis- and trans- side of Golgi stacks, with the membrane system requiring high flexibility and fast self-closing of the involved membranes. These properties may—possibly partially—be controlled by endogenous hydrophobic membrane “fluidizers” for which FLSs are prime candidates. A recent reexamination of unexplained classical data suggests that they are likely synthesized by the Golgi itself. This game-changing hypothesis is endorsed by several arguments and data, some of which date from 1964, that the insect corpus allatum (CA), which is the major production site of farnesol-esters, has active Golgi systems. Thus, in addition to secreting FLS, in particular juvenile hormone(s), it also secretes a protein(s) or peptide(s) with thus far unknown function. This paper suggests answers to various open questions in cell physiology and general endocrinology.
Collapse
Affiliation(s)
- Arnold De Loof
- Research Group of Functional Genomics and Proteomics, Department of Biology, KU Leuven, Leuven, Belgium
| | - Liliane Schoofs
- Research Group of Functional Genomics and Proteomics, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
57
|
Sun Y, Nascimento Da Conceicao V, Ahamad N, Madesh M, Singh BB. Spatial localization of SOCE channels and its modulators regulate neuronal physiology and contributes to pathology. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
58
|
Bruce JIE, James AD. Targeting the Calcium Signalling Machinery in Cancer. Cancers (Basel) 2020; 12:cancers12092351. [PMID: 32825277 PMCID: PMC7565467 DOI: 10.3390/cancers12092351] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/30/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is caused by excessive cell proliferation and a propensity to avoid cell death, while the spread of cancer is facilitated by enhanced cellular migration, invasion, and vascularization. Cytosolic Ca2+ is central to each of these important processes, yet to date, there are no cancer drugs currently being used clinically, and very few undergoing clinical trials, that target the Ca2+ signalling machinery. The aim of this review is to highlight some of the emerging evidence that targeting key components of the Ca2+ signalling machinery represents a novel and relatively untapped therapeutic strategy for the treatment of cancer.
Collapse
Affiliation(s)
- Jason I. E. Bruce
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Correspondence: ; Tel.: +44-(0)-161-275-5484
| | - Andrew D. James
- Department of Biology, University of York, Heslington, York YO10 5DD, UK;
| |
Collapse
|
59
|
Selenoprotein N is an endoplasmic reticulum calcium sensor that links luminal calcium levels to a redox activity. Proc Natl Acad Sci U S A 2020; 117:21288-21298. [PMID: 32817544 PMCID: PMC7474598 DOI: 10.1073/pnas.2003847117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Selenoprotein N (SEPN1) is a type II transmembrane protein of the endoplasmic reticulum (ER) that senses luminal calcium through an EF-hand domain. On calcium depletion, a SEPN1 oligomer, prevalent under basal calcium concentrations, dissociates to generate a monomeric polypeptide that has enhanced redox trapping potential for its target the calcium pump, SERCA2, as well as for many additional interactors, indicating enhanced reductase activity. Our studies not only support that SEPN1 is one of the long-sought reductases of the ER, but also identify a feedback mechanism through which SEPN1 senses the luminal calcium level to modulate downstream signal transduction. Our results suggest that SEPN1 regulates the SERCA-mediated replenishment of ER calcium stores, a crucial mechanism for excitation-contraction coupling in skeletal muscle. The endoplasmic reticulum (ER) is the reservoir for calcium in cells. Luminal calcium levels are determined by calcium-sensing proteins that trigger calcium dynamics in response to calcium fluctuations. Here we report that Selenoprotein N (SEPN1) is a type II transmembrane protein that senses ER calcium fluctuations by binding this ion through a luminal EF-hand domain. In vitro and in vivo experiments show that via this domain, SEPN1 responds to diminished luminal calcium levels, dynamically changing its oligomeric state and enhancing its redox-dependent interaction with cellular partners, including the ER calcium pump sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). Importantly, single amino acid substitutions in the EF-hand domain of SEPN1 identified as clinical variations are shown to impair its calcium-binding and calcium-dependent structural changes, suggesting a key role of the EF-hand domain in SEPN1 function. In conclusion, SEPN1 is a ER calcium sensor that responds to luminal calcium depletion, changing its oligomeric state and acting as a reductase to refill ER calcium stores.
Collapse
|
60
|
Foulquier F, Legrand D. Biometals and glycosylation in humans: Congenital disorders of glycosylation shed lights into the crucial role of Golgi manganese homeostasis. Biochim Biophys Acta Gen Subj 2020; 1864:129674. [PMID: 32599014 DOI: 10.1016/j.bbagen.2020.129674] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022]
Abstract
About half of the eukaryotic proteins bind biometals that participate in their structure and functions in virtually all physiological processes, including glycosylation. After reviewing the biological roles and transport mechanisms of calcium, magnesium, manganese, zinc and cobalt acting as cofactors of the metalloproteins involved in sugar metabolism and/or glycosylation, the paper will outline the pathologies resulting from a dysregulation of these metals homeostasis and more particularly Congenital Disorders of Glycosylation (CDGs) caused by ion transporter defects. Highlighting of CDGs due to defects in SLC39A8 (ZIP8) and TMEM165, two proteins transporting manganese from the extracellular space to cytosol and from cytosol to the Golgi lumen, respectively, has emphasized the importance of manganese homeostasis for glycosylation. Based on our current knowledge of TMEM165 structure and functions, this review will draw a picture of known and putative mechanisms regulating manganese homeostasis in the secretory pathway.
Collapse
Affiliation(s)
- François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France.
| |
Collapse
|
61
|
Channels that Cooperate with TRPV4 in the Brain. J Mol Neurosci 2020; 70:1812-1820. [PMID: 32524421 DOI: 10.1007/s12031-020-01574-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective Ca2+-permeable cation channel that is a member of the TRP channel family. It is clear that TRPV4 channels are broadly expressed in the brain. As they are expressed on the plasma membrane, they interact with other channels and play a crucial role in nervous system activity. Under some pathological conditions, TRPV4 channels are upregulated and sensitized via cellular signaling pathways, and this can cause nervous system diseases. In this review, we focus on receptors that cooperate with TRPV4, including large-conductance Ca2+-activated K+(BKca) channels, N-methyl-D-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptors (AMPARs), inositol 1,4,5-trisphosphate receptors (IP3Rs), ryanodine receptors (RyRs), aquaporin 4 (AQP4), and other potential cooperative receptors in the brain. The data demonstrate how these channels work together to cause nervous system diseases under pathological conditions. The aim of this review was to discuss the receptors and signaling pathways related to TRPV4 based on recent data on the important physiological functions of TRPV4 channels to provide new clues for future studies and prospective therapeutic targets for related brain diseases.
Collapse
|
62
|
Rawat A, Singh P, Jyoti A, Kaushik S, Srivastava VK. Averting transmission: A pivotal target to manage amoebiasis. Chem Biol Drug Des 2020; 96:731-744. [PMID: 32356312 DOI: 10.1111/cbdd.13699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/18/2022]
Abstract
Amoebiasis is a parasitic infectious disease caused by the enteric protozoan Entamoeba histolytica, a leading basis of deaths accounted to parasites, succeeding malaria and schistosomiasis. Conventional treatment methodologies used to deal with amoebiasis mainly rely on the administration of anti-amoebic compounds and vaccines but are often linked with substantial side-effects on the patient. Besides, cases of development of drug resistance in protozoans have been recorded, contributing further to the reduction in the efficiency of the treatment. Loopholes in the efficacious management of the disease call for the development of novel methodologies to manage amoebiasis. A way to achieve this is by targeting the essential metabolic processes of 'encystation' and 'excystation', and the associated biomolecules, thus interrupting the biphasic life cycle of the parasite. Technologies like the CRISPR-Cas9 system can efficiently be exploited to discover novel and essential molecules that regulate the protozoan's metabolism, while efficiently manipulating and managing the known drug targets, leading to an effective halt and forestall to the enteric infection. This review presents a perspective on these essential metabolic processes and the associated molecules that can be targeted efficaciously to prevent the transmission of amoebiasis, thus managing the disease and proving to be a fruitful endeavour.
Collapse
Affiliation(s)
- Aadish Rawat
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Parikshit Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Anupam Jyoti
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Sanket Kaushik
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | | |
Collapse
|
63
|
Papp B, Launay S, Gélébart P, Arbabian A, Enyedi A, Brouland JP, Carosella ED, Adle-Biassette H. Endoplasmic Reticulum Calcium Pumps and Tumor Cell Differentiation. Int J Mol Sci 2020; 21:ijms21093351. [PMID: 32397400 PMCID: PMC7247589 DOI: 10.3390/ijms21093351] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 12/21/2022] Open
Abstract
Endoplasmic reticulum (ER) calcium homeostasis plays an essential role in cellular calcium signaling, intra-ER protein chaperoning and maturation, as well as in the interaction of the ER with other organelles. Calcium is accumulated in the ER by sarco/endoplasmic reticulum calcium ATPases (SERCA enzymes) that generate by active, ATP-dependent transport, a several thousand-fold calcium ion concentration gradient between the cytosol (low nanomolar) and the ER lumen (high micromolar). SERCA enzymes are coded by three genes that by alternative splicing give rise to several isoforms, which can display isoform-specific calcium transport characteristics. SERCA expression levels and isoenzyme composition vary according to cell type, and this constitutes a mechanism whereby ER calcium homeostasis is adapted to the signaling and metabolic needs of the cell, depending on its phenotype, its state of activation and differentiation. As reviewed here, in several normal epithelial cell types including bronchial, mammary, gastric, colonic and choroid plexus epithelium, as well as in mature cells of hematopoietic origin such as pumps are simultaneously expressed, whereas in corresponding tumors and leukemias SERCA3 expression is selectively down-regulated. SERCA3 expression is restored during the pharmacologically induced differentiation of various cancer and leukemia cell types. SERCA3 is a useful marker for the study of cell differentiation, and the loss of SERCA3 expression constitutes a previously unrecognized example of the remodeling of calcium homeostasis in tumors.
Collapse
Affiliation(s)
- Bela Papp
- Institut National de la Santé et de la Recherche Médicale, UMR U976, Institut Saint-Louis, 75010 Paris, France
- Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Université de Paris, 75010 Paris, France
- CEA, DRF-Institut Francois Jacob, Department of Hemato-Immunology Research, Hôpital Saint-Louis, 75010 Paris, France;
- Correspondence: or
| | - Sophie Launay
- EA481, UFR Santé, Université de Bourgogne Franche-Comté, 25000 Besançon, France;
| | - Pascal Gélébart
- Department of Clinical Science-Hematology Section, Haukeland University Hospital, University of Bergen, 5021 Bergen, Norway;
| | - Atousa Arbabian
- Laboratoire d’Innovation Vaccins, Institut Pasteur de Paris, 75015 Paris, France;
| | - Agnes Enyedi
- Second Department of Pathology, Semmelweis University, 1091 Budapest, Hungary;
| | - Jean-Philippe Brouland
- Institut Universitaire de Pathologie, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland;
| | - Edgardo D. Carosella
- CEA, DRF-Institut Francois Jacob, Department of Hemato-Immunology Research, Hôpital Saint-Louis, 75010 Paris, France;
| | - Homa Adle-Biassette
- AP-HP, Service d’Anatomie et Cytologie Pathologiques, Hôpital Lariboisière, 75010 Paris, France;
- Université de Paris, NeuroDiderot, Inserm UMR 1141, 75019 Paris, France
| |
Collapse
|
64
|
Wang X, Mick G, McCormick K. Pyridine nucleotide regulation of hepatic endoplasmic reticulum calcium uptake. Physiol Rep 2020; 7:e14151. [PMID: 31222964 PMCID: PMC6586769 DOI: 10.14814/phy2.14151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/31/2019] [Indexed: 01/03/2023] Open
Abstract
Pyridine nucleotides serve an array of intracellular metabolic functions such as, to name a few, shuttling electrons in enzymatic reactions, safeguarding the redox state against reactive oxygen species, cytochrome P450 (CYP) enzyme detoxification pathways and, relevant to this study, the regulation of ion fluxes. In particular, the maintenance of a steep calcium gradient between the cytosol and endoplasmic reticulum (ER), without which apoptosis ensues, is achieved by an elaborate combination of energy–requiring ER membrane pumps and efflux channels. In liver microsomes, net calcium uptake was inhibited by physiological concentrations of NADP. In the presence of 1 mmol/L NADP, calcium uptake was attenuated by nearly 80%, additionally, this inhibitory effect was blunted by concomitant addition of NADPH. No other nicotinamide containing compounds ‐save a slight inhibition by NAADP‐hindered calcium uptake; thus, only oxidized pyridine nucleotides, or related compounds with a phosphate moiety, had an imposing effect. Moreover, the NADP inhibition was evident even after selectively blocking ER calcium efflux channels. Given the fundamental role of endoplasmic calcium homeostasis, it is plausible that changes in cytosolic NADP concentration, for example, during anabolic processes, could regulate net ER calcium uptake.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gail Mick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kenneth McCormick
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
65
|
Chen J, Sitsel A, Benoy V, Sepúlveda MR, Vangheluwe P. Primary Active Ca 2+ Transport Systems in Health and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035113. [PMID: 31501194 DOI: 10.1101/cshperspect.a035113] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calcium ions (Ca2+) are prominent cell signaling effectors that regulate a wide variety of cellular processes. Among the different players in Ca2+ homeostasis, primary active Ca2+ transporters are responsible for keeping low basal Ca2+ levels in the cytosol while establishing steep Ca2+ gradients across intracellular membranes or the plasma membrane. This review summarizes our current knowledge on the three types of primary active Ca2+-ATPases: the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pumps, the secretory pathway Ca2+- ATPase (SPCA) isoforms, and the plasma membrane Ca2+-ATPase (PMCA) Ca2+-transporters. We first discuss the Ca2+ transport mechanism of SERCA1a, which serves as a reference to describe the Ca2+ transport of other Ca2+ pumps. We further highlight the common and unique features of each isoform and review their structure-function relationship, expression pattern, regulatory mechanisms, and specific physiological roles. Finally, we discuss the increasing genetic and in vivo evidence that links the dysfunction of specific Ca2+-ATPase isoforms to a broad range of human pathologies, and highlight emerging therapeutic strategies that target Ca2+ pumps.
Collapse
Affiliation(s)
- Jialin Chen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Aljona Sitsel
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Veronick Benoy
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, 18071 Granada, Spain
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
66
|
Abstract
Ionized calcium (Ca2+) is the most versatile cellular messenger. All cells use Ca2+ signals to regulate their activities in response to extrinsic and intrinsic stimuli. Alterations in cellular Ca2+ signaling and/or Ca2+ homeostasis can subvert physiological processes into driving pathological outcomes. Imaging of living cells over the past decades has demonstrated that Ca2+ signals encode information in their frequency, kinetics, amplitude, and spatial extent. These parameters alter depending on the type and intensity of stimulation, and cellular context. Moreover, it is evident that different cell types produce widely varying Ca2+ signals, with properties that suit their physiological functions. This primer discusses basic principles and mechanisms underlying cellular Ca2+ signaling and Ca2+ homeostasis. Consequently, we have cited some historical articles in addition to more recent findings. A brief summary of the core features of cellular Ca2+ signaling is provided, with particular focus on Ca2+ stores and Ca2+ transport across cellular membranes, as well as mechanisms by which Ca2+ signals activate downstream effector systems.
Collapse
|
67
|
Wang WA, Agellon LB, Michalak M. Organellar Calcium Handling in the Cellular Reticular Network. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a038265. [PMID: 31358518 DOI: 10.1101/cshperspect.a038265] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ca2+ is an important intracellular messenger affecting diverse cellular processes. In eukaryotic cells, Ca2+ is handled by a myriad of Ca2+-binding proteins found in organelles that are organized into the cellular reticular network (CRN). The network is comprised of the endoplasmic reticulum, Golgi apparatus, lysosomes, membranous components of the endocytic and exocytic pathways, peroxisomes, and the nuclear envelope. Membrane contact sites between the different components of the CRN enable the rapid movement of Ca2+, and communication of Ca2+ status, within the network. Ca2+-handling proteins that reside in the CRN facilitate Ca2+ sensing, buffering, and cellular signaling to coordinate the many processes that operate within the cell.
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2S7, Canada
| |
Collapse
|
68
|
Chan H, Bhide KP, Vaidyam A, Hedrick V, Sobreira TJP, Sors TG, Grant RW, Aryal UK. Proteomic Analysis of 3T3-L1 Adipocytes Treated with Insulin and TNF-α. Proteomes 2019; 7:35. [PMID: 31635166 PMCID: PMC6958341 DOI: 10.3390/proteomes7040035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance is an indication of early stage Type 2 diabetes (T2D). Insulin resistant adipose tissues contain higher levels of insulin than the physiological level, as well as higher amounts of intracellular tumor necrosis factor-α (TNF-α) and other cytokines. However, the mechanism of insulin resistance remains poorly understood. To better understand the roles played by insulin and TNF-α in insulin resistance, we performed proteomic analysis of differentiated 3T3-L1 adipocytes treated with insulin (Ins), TNF-α (TNF), and both (Ins + TNF). Out of the 693 proteins identified, the abundances of 78 proteins were significantly different (p < 0.05). Carnitine parmitoyltransferase-2 (CPT2), acetyl CoA carboxylase 1 (ACCAC-1), ethylmalonyl CoA decarboxylase (ECHD1), and methylmalonyl CoA isomerase (MCEE), enzymes required for fatty acid β-oxidation and respiratory electron transport, and β-glucuronidase, an enzyme responsible for the breakdown of complex carbohydrates, were down-regulated in all the treatment groups, compared to the control group. In contrast, superoxide dismutase 2 (SOD2), protein disulfide isomerase (PDI), and glutathione reductase, which are the proteins responsible for cytoskeletal structure, protein folding, degradation, and oxidative stress responses, were up-regulated. This suggests higher oxidative stress in cells treated with Ins, TNF, or both. We proposed a conceptual metabolic pathway impacted by the treatments and their possible link to insulin resistance or T2D.
Collapse
Affiliation(s)
- Hayley Chan
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Ketaki P Bhide
- College of Agriculture, Purdue University, West Lafayette, IN 47907, USA.
| | - Aditya Vaidyam
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Victoria Hedrick
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
| | | | - Thomas G Sors
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| | - Ryan W Grant
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Uma K Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
69
|
Vamecq J, Papegay B, Nuyens V, Boogaerts J, Leo O, Kruys V. Mitochondrial dysfunction, AMPK activation and peroxisomal metabolism: A coherent scenario for non-canonical 3-methylglutaconic acidurias. Biochimie 2019; 168:53-82. [PMID: 31626852 DOI: 10.1016/j.biochi.2019.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
The occurrence of 3-methylglutaconic aciduria (3-MGA) is a well understood phenomenon in leucine oxidation and ketogenesis disorders (primary 3-MGAs). In contrast, its genesis in non-canonical (secondary) 3-MGAs, a growing-up group of disorders encompassing more than a dozen of inherited metabolic diseases, is a mystery still remaining unresolved for three decades. To puzzle out this anthologic problem of metabolism, three clues were considered: (i) the variety of disorders suggests a common cellular target at the cross-road of metabolic and signaling pathways, (ii) the response to leucine loading test only discriminative for primary but not secondary 3-MGAs suggests these latter are disorders of extramitochondrial HMG-CoA metabolism as also attested by their failure to increase 3-hydroxyisovalerate, a mitochondrial metabolite accumulating only in primary 3-MGAs, (iii) the peroxisome is an extramitochondrial site possessing its own pool and displaying metabolism of HMG-CoA, suggesting its possible involvement in producing extramitochondrial 3-methylglutaconate (3-MG). Following these clues provides a unifying common basis to non-canonical 3-MGAs: constitutive mitochondrial dysfunction induces AMPK activation which, by inhibiting early steps in cholesterol and fatty acid syntheses, pipelines cytoplasmic acetyl-CoA to peroxisomes where a rise in HMG-CoA followed by local dehydration and hydrolysis may lead to 3-MGA yield. Additional contributors are considered, notably for 3-MGAs associated with hyperammonemia, and to a lesser extent in CLPB deficiency. Metabolic and signaling itineraries followed by the proposed scenario are essentially sketched, being provided with compelling evidence from the literature coming in their support.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, CHU Lille, Univ Lille, Department of Biochemistry and Molecular Biology, Laboratory of Hormonology, Metabolism-Nutrition & Oncology (HMNO), Center of Biology and Pathology (CBP) Pierre-Marie Degand, CHRU Lille, EA 7364 RADEME, University of North France, Lille, France.
| | - Bérengère Papegay
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Vincent Nuyens
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Jean Boogaerts
- Laboratory of Experimental Medicine (ULB unit 222), University Hospital Center, Charleroi, (CHU Charleroi), Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| | - Véronique Kruys
- Laboratory of Molecular Biology of the Gene, Department of Molecular Biology, ULB Immunology Research Center (UIRC), Free University of Brussels (ULB), Gosselies, Belgium
| |
Collapse
|
70
|
Min DJ, Zhao Y, Monks A, Palmisano A, Hose C, Teicher BA, Doroshow JH, Simon RM. Identification of pharmacodynamic biomarkers and common molecular mechanisms of response to genotoxic agents in cancer cell lines. Cancer Chemother Pharmacol 2019; 84:771-780. [PMID: 31367787 PMCID: PMC8127867 DOI: 10.1007/s00280-019-03898-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Genotoxic agents (GAs) including cisplatin, doxorubicin, gemcitabine, and topotecan are often used in cancer treatment. However, the response to GAs is variable among patients and predictive biomarkers are inadequate to select patients for treatment. Accurate and rapid pharmacodynamics measures of response can, thus, be useful for monitoring therapy and improve clinical outcomes. METHODS This study focuses on integrating a database of genome-wide response to treatment (The NCI Transcriptional Pharmacodynamics Workbench) with a database of baseline gene expression (GSE32474) for the NCI-60 cell lines to identify mechanisms of response and pharmacodynamic (PD) biomarkers. RESULTS AND CONCLUSIONS Our analysis suggests that GA-induced endoplasmic reticulum (ER) stress may signal for GA-induced cell death. Reducing the uptake of GA, activating DNA repair, and blocking ER-stress induction cooperate to prevent GA-induced cell death in the GA-resistant cells. ATF3, DDIT3, CARS, and PPP1R15A appear as possible candidate PD biomarkers for monitoring the progress of GA treatment. Further validation studies on the proposed intrinsic drug-resistant mechanism and candidate genes are needed using in vivo data from either patient-derived xenograft models or clinical chemotherapy trials.
Collapse
Affiliation(s)
- Dong-Joon Min
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Yingdong Zhao
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Anne Monks
- Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Alida Palmisano
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA
| | - Curtis Hose
- Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Beverly A Teicher
- Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 20892, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Richard M Simon
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD, 20850, USA.
| |
Collapse
|
71
|
Karnkowska A, Treitli SC, Brzoň O, Novák L, Vacek V, Soukal P, Barlow LD, Herman EK, Pipaliya SV, Pánek T, Žihala D, Petrželková R, Butenko A, Eme L, Stairs CW, Roger AJ, Eliáš M, Dacks JB, Hampl V. The Oxymonad Genome Displays Canonical Eukaryotic Complexity in the Absence of a Mitochondrion. Mol Biol Evol 2019; 36:2292-2312. [PMID: 31387118 PMCID: PMC6759080 DOI: 10.1093/molbev/msz147] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The discovery that the protist Monocercomonoides exilis completely lacks mitochondria demonstrates that these organelles are not absolutely essential to eukaryotic cells. However, the degree to which the metabolism and cellular systems of this organism have adapted to the loss of mitochondria is unknown. Here, we report an extensive analysis of the M. exilis genome to address this question. Unexpectedly, we find that M. exilis genome structure and content is similar in complexity to other eukaryotes and less "reduced" than genomes of some other protists from the Metamonada group to which it belongs. Furthermore, the predicted cytoskeletal systems, the organization of endomembrane systems, and biosynthetic pathways also display canonical eukaryotic complexity. The only apparent preadaptation that permitted the loss of mitochondria was the acquisition of the SUF system for Fe-S cluster assembly and the loss of glycine cleavage system. Changes in other systems, including in amino acid metabolism and oxidative stress response, were coincident with the loss of mitochondria but are likely adaptations to the microaerophilic and endobiotic niche rather than the mitochondrial loss per se. Apart from the lack of mitochondria and peroxisomes, we show that M. exilis is a fully elaborated eukaryotic cell that is a promising model system in which eukaryotic cell biology can be investigated in the absence of mitochondria.
Collapse
Affiliation(s)
- Anna Karnkowska
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
- Department of Molecular Phylogenetics and Evolution, Faculty of Biology, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Sebastian C Treitli
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Ondřej Brzoň
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Lukáš Novák
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Vojtěch Vacek
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Petr Soukal
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Lael D Barlow
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Emily K Herman
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Shweta V Pipaliya
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Tomáš Pánek
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - David Žihala
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Romana Petrželková
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Anzhelika Butenko
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Laura Eme
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Courtney W Stairs
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Andrew J Roger
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Canada
| | - Marek Eliáš
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
- Institute of Environmental Technologies, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Joel B Dacks
- Division of Infectious Disease, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Vladimír Hampl
- Department of Parasitology, BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| |
Collapse
|
72
|
McDonnell SJ, Spiller DG, White MRH, Prior IA, Paraoan L. ER stress-linked autophagy stabilizes apoptosis effector PERP and triggers its co-localization with SERCA2b at ER-plasma membrane junctions. Cell Death Discov 2019; 5:132. [PMID: 31508245 PMCID: PMC6718399 DOI: 10.1038/s41420-019-0212-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 01/01/2023] Open
Abstract
Specific molecular interactions that underpin the switch between ER stress-triggered autophagy-mediated cellular repair and cellular death by apoptosis are not characterized. This study reports the unexpected interaction elicited by ER stress between the plasma membrane (PM)-localized apoptosis effector PERP and the ER Ca2+ pump SERCA2b. We show that the p53 effector PERP, which specifically induces apoptosis when expressed above a threshold level, has a heterogeneous distribution across the PM of un-stressed cells and is actively turned over by the lysosome. PERP is upregulated following sustained starvation-induced autophagy, which precedes the onset of apoptosis indicating that PERP protein levels are controlled by a lysosomal pathway that is sensitive to cellular physiological state. Furthermore, ER stress stabilizes PERP at the PM and induces its increasing co-localization with SERCA2b at ER–PM junctions. The findings highlight a novel crosstalk between pro-survival autophagy and pro-death apoptosis pathways and identify, for the first time, accumulation of an apoptosis effector to ER–PM junctions in response to ER stress.
Collapse
Affiliation(s)
- Samantha J McDonnell
- 1Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX UK
| | - David G Spiller
- 2Systems Microscopy Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
| | - Michael R H White
- 3School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
| | - Ian A Prior
- 4Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX UK
| | - Luminita Paraoan
- 1Department of Eye and Vision Science, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX UK
| |
Collapse
|
73
|
ER Ca 2+ release and store-operated Ca 2+ entry - partners in crime or independent actors in oncogenic transformation? Cell Calcium 2019; 82:102061. [PMID: 31394337 DOI: 10.1016/j.ceca.2019.102061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Ca2+ is a pleiotropic messenger that controls life and death decisions from fertilisation until death. Cellular Ca2+ handling mechanisms show plasticity and are remodelled throughout life to meet the changing needs of the cell. In turn, as the demands on a cell alter, for example through a change in its niche environment or its functional requirements, Ca2+ handling systems may be targeted to sustain the remodelled cellular state. Nowhere is this more apparent than in cancer. Oncogenic transformation is a multi-stage process during which normal cells become progressively differentiated towards a cancerous state that is principally associated with enhanced proliferation and avoidance of death. Ca2+ signalling is intimately involved in almost all aspects of the life of a transformed cell and alterations in Ca2+ handling have been observed in cancer. Moreover, this remodelling of Ca2+ signalling pathways is also required in some cases to sustain the transformed phenotype. As such, Ca2+ handling is hijacked by oncogenic processes to deliver and maintain the transformed phenotype. Central to generation of intracellular Ca2+ signals is the release of Ca2+ from the endoplasmic reticulum intracellular (ER) Ca2+ store via inositol 1,4,5-trisphosphate receptors (InsP3Rs). Upon depletion of ER Ca2+, store-operated Ca2+ entry (SOCE) across the plasma membrane occurs via STIM-gated Orai channels. SOCE serves to both replenish stores but also sustain Ca2+ signalling events. Here, we will discuss the role and regulation of these two signalling pathways and their interplay in oncogenic transformation.
Collapse
|
74
|
Lee JH, Han JH, Kim H, Park SM, Joe EH, Jou I. Parkinson's disease-associated LRRK2-G2019S mutant acts through regulation of SERCA activity to control ER stress in astrocytes. Acta Neuropathol Commun 2019; 7:68. [PMID: 31046837 PMCID: PMC6498585 DOI: 10.1186/s40478-019-0716-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 04/07/2019] [Indexed: 01/10/2023] Open
Abstract
Accumulating evidence indicates that endoplasmic reticulum (ER) stress is a common feature of Parkinson’s disease (PD) and further suggests that several PD-related genes are responsible for ER dysfunction. However, the underlying mechanisms are largely unknown. Here, we defined the mechanism by which LRRK2-G2019S (LRRK2-GS), a pathogenic mutation in the PD-associated gene LRRK2, accelerates ER stress and cell death. Treatment of cells with α-synuclein increased the expression of ER stress proteins and subsequent cell death in LRRK2-GS astrocytes. Intriguingly, we found that LRRK2-GS localizes to the ER membrane, where it interacts with sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) and suppress its activity by preventing displacement of phospholamban (PLN). LRRK2-GS–mediated SERCA malfunction leads to ER Ca2+ depletion, which induces the formation of mitochondria-ER contacts and subsequent Ca2+ overload in mitochondria, ultimately resulting in mitochondrial dysfunction. Collectively, our data suggest that, in astrocytes, LRRK2-GS impairs ER Ca2+ homeostasis, which determines cell survival, and as a result, could contribute to the development of PD.
Collapse
|
75
|
Eckstein M, Vaeth M, Aulestia FJ, Costiniti V, Kassam SN, Bromage TG, Pedersen P, Issekutz T, Idaghdour Y, Moursi AM, Feske S, Lacruz RS. Differential regulation of Ca 2+ influx by ORAI channels mediates enamel mineralization. Sci Signal 2019; 12:12/578/eaav4663. [PMID: 31015290 DOI: 10.1126/scisignal.aav4663] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Store-operated Ca2+ entry (SOCE) channels are highly selective Ca2+ channels activated by the endoplasmic reticulum (ER) sensors STIM1 and STIM2. Their direct interaction with the pore-forming plasma membrane ORAI proteins (ORAI1, ORAI2, and ORAI3) leads to sustained Ca2+ fluxes that are critical for many cellular functions. Mutations in the human ORAI1 gene result in immunodeficiency, anhidrotic ectodermal dysplasia, and enamel defects. In our investigation of the role of ORAI proteins in enamel, we identified enamel defects in a patient with an ORAI1 null mutation. Targeted deletion of the Orai1 gene in mice showed enamel defects and reduced SOCE in isolated enamel cells. However, Orai2-/- mice showed normal enamel despite having increased SOCE in the enamel cells. Knockdown experiments in the enamel cell line LS8 suggested that ORAI2 and ORAI3 modulated ORAI1 function, with ORAI1 and ORAI2 being the main contributors to SOCE. ORAI1-deficient LS8 cells showed altered mitochondrial respiration with increased oxygen consumption rate and ATP, which was associated with altered redox status and enhanced ER Ca2+ uptake, likely due to S-glutathionylation of SERCA pumps. Our findings demonstrate an important role of ORAI1 in Ca2+ influx in enamel cells and establish a link between SOCE, mitochondrial function, and redox homeostasis.
Collapse
Affiliation(s)
- Miriam Eckstein
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Francisco J Aulestia
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Veronica Costiniti
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA
| | - Serena N Kassam
- Department of Pediatric Dentistry, New York University College of Dentistry, New York, NY 10010, USA
| | - Timothy G Bromage
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA.,Department of Biomaterials, New York University College of Dentistry, New York, NY 10010, USA
| | - Pal Pedersen
- Carl Zeiss Microscopy, LLC, Thornwood, NY 10594, USA
| | - Thomas Issekutz
- Department of Pediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Youssef Idaghdour
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amr M Moursi
- Department of Pediatric Dentistry, New York University College of Dentistry, New York, NY 10010, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA.
| |
Collapse
|
76
|
Xia M, Zhang Y, Jin K, Lu Z, Zeng Z, Xiong W. Communication between mitochondria and other organelles: a brand-new perspective on mitochondria in cancer. Cell Biosci 2019; 9:27. [PMID: 30931098 PMCID: PMC6425566 DOI: 10.1186/s13578-019-0289-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/09/2019] [Indexed: 12/24/2022] Open
Abstract
Mitochondria are energy factories of cells and are important pivots for intracellular interactions with other organelles. They interact with the endoplasmic reticulum, peroxisomes, and nucleus through signal transduction, vesicle transport, and membrane contact sites to regulate energy metabolism, biosynthesis, immune response, and cell turnover. However, when the communication between organelles fails and the mitochondria are dysfunctional, it may induce tumorigenesis. In this review, we elaborate on how mitochondria interact with the endoplasmic reticulum, peroxisomes, and cell nuclei, as well as the relation between organelle communication and tumor development .
Collapse
Affiliation(s)
- MengFang Xia
- 1NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan China.,2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China.,3Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan China
| | - YaZhuo Zhang
- 1NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan China.,2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China.,3Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Ke Jin
- 2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China
| | - ZiTong Lu
- 2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China
| | - Zhaoyang Zeng
- 1NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan China.,2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China.,3Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Wei Xiong
- 1NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan China.,2The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan China.,3Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan China
| |
Collapse
|
77
|
Sitsel A, De Raeymaecker J, Drachmann ND, Derua R, Smaardijk S, Andersen JL, Vandecaetsbeek I, Chen J, De Maeyer M, Waelkens E, Olesen C, Vangheluwe P, Nissen P. Structures of the heart specific SERCA2a Ca 2+-ATPase. EMBO J 2019; 38:embj.2018100020. [PMID: 30777856 DOI: 10.15252/embj.2018100020] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 12/29/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
The sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2a (SERCA2a) performs active reuptake of cytoplasmic Ca2+ and is a major regulator of cardiac muscle contractility. Dysfunction or dysregulation of SERCA2a is associated with heart failure, while restoring its function is considered as a therapeutic strategy to restore cardiac performance. However, its structure has not yet been determined. Based on native, active protein purified from pig ventricular muscle, we present the first crystal structures of SERCA2a, determined in the CPA-stabilized E2-AlF4- form (3.3 Å) and the Ca2+-occluded [Ca2]E1-AMPPCP form (4.0 Å). The structures are similar to the skeletal muscle isoform SERCA1a pointing to a conserved mechanism. We seek to explain the kinetic differences between SERCA1a and SERCA2a. We find that several isoform-specific residues are acceptor sites for post-translational modifications. In addition, molecular dynamics simulations predict that isoform-specific residues support distinct intramolecular interactions in SERCA2a and SERCA1a. Our experimental observations further indicate that isoform-specific intramolecular interactions are functionally relevant, and may explain the kinetic differences between SERCA2a and SERCA1a.
Collapse
Affiliation(s)
- Aljona Sitsel
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,Center for Membrane Proteins in Cells and Disease - PUMPkin, Danish National Research Foundation, Aarhus C, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
| | | | - Nikolaj Düring Drachmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,Center for Membrane Proteins in Cells and Disease - PUMPkin, Danish National Research Foundation, Aarhus C, Denmark
| | - Rita Derua
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,SyBioMa, KU Leuven, Leuven, Belgium
| | - Susanne Smaardijk
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jacob Lauwring Andersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,Center for Membrane Proteins in Cells and Disease - PUMPkin, Danish National Research Foundation, Aarhus C, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Jialin Chen
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | | | - Etienne Waelkens
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,SyBioMa, KU Leuven, Leuven, Belgium
| | - Claus Olesen
- Center for Membrane Proteins in Cells and Disease - PUMPkin, Danish National Research Foundation, Aarhus C, Denmark .,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Peter Vangheluwe
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Poul Nissen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark .,Center for Membrane Proteins in Cells and Disease - PUMPkin, Danish National Research Foundation, Aarhus C, Denmark.,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
| |
Collapse
|
78
|
Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors. Cell Death Dis 2019; 10:128. [PMID: 30755589 PMCID: PMC6372664 DOI: 10.1038/s41419-019-1413-8] [Citation(s) in RCA: 840] [Impact Index Per Article: 140.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022]
Abstract
The NLRP3 inflammasome is a multimeric protein complex that initiates an inflammatory form of cell death and triggers the release of proinflammatory cytokines IL-1β and IL-18. The NLRP3 inflammasome has been implicated in a wide range of diseases, including Alzheimer’s disease, Prion diseases, type 2 diabetes, and some infectious diseases. It has been found that a variety of stimuli including danger-associated molecular patterns (DAMPs, such as silica and uric acid crystals) and pathogen-associated molecular patterns (PAMPs) can activate NLRP3 inflammasome, but the specific regulatory mechanisms of NLRP3 inflammasome activation remain unclear. Understanding the mechanisms of NLRP3 activation will enable the development of its specific inhibitors to treat NLRP3-related diseases. In this review, we summarize current understanding of the regulatory mechanisms of NLRP3 inflammasome activation as well as inhibitors that specifically and directly target NLRP3.
Collapse
|
79
|
Abstract
Naturally occurring mutations of a calcium ion transporter can cause a skin condition known as Darier's disease. In this issue of JBC, Mikkelsen et al. describe a particularly interesting Darier's mutation that alters calcium transport by disrupting a kinetic braking mechanism that is unique to the SERCA2b calcium pump isoform. The study provides new insight into the intrinsic regulation of this transporter and reveals how disruption of regulation can lead to disease in Darier's patients.
Collapse
Affiliation(s)
- Seth L Robia
- From the Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois 60153 and
| | - Howard S Young
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
80
|
De Loof A, Schoofs L. Intraluminal Farnesol and Farnesal in the Mealworm's Alimentary Canal: An Unusual Storage Site Uncovering Hidden Eukaryote Ca 2+-Homeostasis-Dependent "Golgicrine" Activities. Front Endocrinol (Lausanne) 2019; 10:885. [PMID: 31920991 PMCID: PMC6930878 DOI: 10.3389/fendo.2019.00885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Farnesol, the sesquiterpenoid precursor of the six presently known insect juvenile hormones (JHs) was for the first time chemically identified in 1961, not in JH synthesizing glands or whole body extracts, but in excrements of the mealworm Tenebrio molitor. This finding was thought to be irrelevant and remained unexplored. In 1970, it was reported that the fall to zero of the JH titer in both prediapausing adults and in last instar larvae of the Colorado potato beetle causes severe malfunctioning of the Golgi system in the fat body, among various other effects. This endomembrane system in the cytoplasm resides at the intersection of the secretory, lysosomal, and endocytic pathways and is required for the processing of secretory proteins. Why the Golgi needs farnesol-like endogenous sesquiterpenoids (FLS) for its proper functioning has also never been further investigated. In 1999, farnesol was found to be a natural endogenous ligand for particular types of voltage-gated Ca2+ channels in mammalian cells, a finding that also remained undervalued. Only since 2014 more attention has been paid to the functional research of the "noble unknown" farnesol, in particular to its Ca2+-homeostasis-related juvenilizing and anti-apoptotic activities. Here, we introduce the term "Golgicrine activity" that addresses the secretory activity of the RER-Golgi system from its role in Ca2+-homeostasis rather than from its conventional role in mere protein secretion. Golgicrine activity attributes the so far forgotten role of farnesol-like sesquiterpenoids in proper Golgi functioning, and unites the endocrine, exocrine and enterocrine functions of these sesquiterpenoids. This out of the box view may open novel perspectives for the better understanding of particular inflammatory bowel diseases and of neurodegenerative diseases as well, because the early initiation of Alzheimer's disease may possibly result from malfunctioning of the mevalonate-farnesol-cholesterol biosynthetic pathway and thus might be a farnesol- and Ca2+-homeostasis-dependent Golgicrine issue.
Collapse
|
81
|
Narayanaswamy N, Chakraborty K, Saminathan A, Zeichner E, Leung K, Devany J, Krishnan Y. A pH-correctable, DNA-based fluorescent reporter for organellar calcium. Nat Methods 2018; 16:95-102. [PMID: 30532082 DOI: 10.1038/s41592-018-0232-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/30/2018] [Indexed: 01/01/2023]
Abstract
It is extremely challenging to quantitate lumenal Ca2+ in acidic Ca2+ stores of the cell because all Ca2+ indicators are pH sensitive, and Ca2+ transport is coupled to pH in acidic organelles. We have developed a fluorescent DNA-based reporter, CalipHluor, that is targetable to specific organelles. By ratiometrically reporting lumenal pH and Ca2+ simultaneously, CalipHluor functions as a pH-correctable Ca2+ reporter. By targeting CalipHluor to the endolysosomal pathway, we mapped lumenal Ca2+ changes during endosomal maturation and found a surge in lumenal Ca2+ specifically in lysosomes. Using lysosomal proteomics and genetic analysis, we found that catp-6, a Caenorhabditis elegans homolog of ATP13A2, was responsible for lysosomal Ca2+ accumulation-an example of a lysosome-specific Ca2+ importer in animals. By enabling the facile quantification of compartmentalized Ca2+, CalipHluor can expand the understanding of subcellular Ca2+ importers.
Collapse
Affiliation(s)
- Nagarjun Narayanaswamy
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
| | - Kasturi Chakraborty
- Department of Chemistry, The University of Chicago, Chicago, IL, USA. .,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA.
| | - Anand Saminathan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
| | | | - KaHo Leung
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA
| | - John Devany
- Department of Physics, The University of Chicago, Chicago, IL, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA. .,Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
82
|
Abstract
In the last few decades, a large body of experimental evidence has highlighted the complex role for mitochondria in eukaryotic cells: they are not only the site of aerobic metabolism (thus providing most of the ATP supply for endergonic processes) but also a crucial checkpoint of cell death processes (both necrosis and apoptosis) and autophagy. For this purpose, mitochondria must receive and decode the wide variety of physiological and pathological stimuli impacting on the cell. The “old” notion that mitochondria possess a sophisticated machinery for accumulating and releasing Ca
2+, the most common and versatile second messenger of eukaryotic cells, is thus no surprise. What may be surprising is that the identification of the molecules involved in mitochondrial Ca
2+ transport occurred only in the last decade for both the influx (the mitochondrial Ca
2+ uniporter, MCU) and the efflux (the sodium calcium exchanger, NCX) pathways. In this review, we will focus on the description of the amazing molecular complexity of the MCU complex, highlighting the numerous functional implications of the tissue-specific expression of the variants of the channel pore components (MCU/MCUb) and of the associated proteins (MICU 1, 2, and 3, EMRE, and MCUR1).
Collapse
Affiliation(s)
- Giorgia Pallafacchina
- Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy.,Italian National Research Council (CNR), Neuroscience Institute, Padua, 35131, Italy
| | - Sofia Zanin
- Department of Medicine, University of Padua, Padua, 35128, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, 35131, Italy
| |
Collapse
|
83
|
Houdou M, Lebredonchel E, Garat A, Duvet S, Legrand D, Decool V, Klein A, Ouzzine M, Gasnier B, Potelle S, Foulquier F. Involvement of thapsigargin- and cyclopiazonic acid-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn 2. FASEB J 2018; 33:2669-2679. [PMID: 30307768 DOI: 10.1096/fj.201800387r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Congenital disorders of glycosylation are severe inherited diseases in which aberrant protein glycosylation is a hallmark. Transmembrane protein 165 (TMEM165) is a novel Golgi transmembrane protein involved in type II congenital disorders of glycosylation. Although its biologic function is still a controversial issue, we have demonstrated that the Golgi glycosylation defect due to TMEM165 deficiency resulted from a Golgi Mn2+ homeostasis defect. The goal of this study was to delineate the cellular pathway by which extracellular Mn2+ rescues N-glycosylation in TMEM165 knockout (KO) cells. We first demonstrated that after extracellular exposure, Mn2+ uptake by HEK293 cells at the plasma membrane did not rely on endocytosis but was likely done by plasma membrane transporters. Second, we showed that the secretory pathway Ca2+-ATPase 1, also known to mediate the influx of cytosolic Mn2+ into the lumen of the Golgi apparatus, is not crucial for the Mn2+-induced rescue glycosylation of lysosomal-associated membrane protein 2 (LAMP2). In contrast, our results demonstrate the involvement of cyclopiazonic acid- and thapsigargin (Tg)-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+. Interestingly, overexpression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) 2b isoform in TMEM165 KO cells partially rescues the observed LAMP2 glycosylation defect. Overall, this study indicates that the rescue of Golgi N-glycosylation defects in TMEM165 KO cells by extracellular Mn2+ involves the activity of Tg and cyclopiazonic acid-sensitive pumps, probably the SERCA pumps.-Houdou, M., Lebredonchel, E., Garat, A., Duvet, S., Legrand, D., Decool, V., Klein, A., Ouzzine, M., Gasnier, B., Potelle, S., Foulquier, F. Involvement of thapsigargin- and cyclopiazonic acid-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+.
Collapse
Affiliation(s)
- Marine Houdou
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Elodie Lebredonchel
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Anne Garat
- Centre Hospitalier Universitaire (CHU) Lille, Institut Pasteur de Lille, Equipe d'Accueil (EA) 4483, Impact de l'Environnement Chimique sur la Santé Humaine (IMPECS), University of Lille, Lille, France
| | - Sandrine Duvet
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Dominique Legrand
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Valérie Decool
- Centre Hospitalier Universitaire (CHU) Lille, Institut Pasteur de Lille, Equipe d'Accueil (EA) 4483, Impact de l'Environnement Chimique sur la Santé Humaine (IMPECS), University of Lille, Lille, France
| | - André Klein
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - Mohamed Ouzzine
- Unité Mixte de Recherche (UMR) 7365, Centre National de la Recherche Scientifique (CNRS)-Université de Lorraine, Biopôle-Faculté de Médecine, Vandoeuvre-lès-Nancy, France
| | - Bruno Gasnier
- Neurophotonics Laboratory, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8250, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Sven Potelle
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| | - François Foulquier
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), University of Lille, Lille, France
| |
Collapse
|
84
|
Roest G, La Rovere RM, Bultynck G, Parys JB. IP 3 Receptor Properties and Function at Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 981:149-178. [PMID: 29594861 DOI: 10.1007/978-3-319-55858-5_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) is a ubiquitously expressed Ca2+-release channel localized in the endoplasmic reticulum (ER). The intracellular Ca2+ signals originating from the activation of the IP3R regulate multiple cellular processes including the control of cell death versus cell survival via their action on apoptosis and autophagy. The exact role of the IP3Rs in these two processes does not only depend on their activity, which is modulated by the cytosolic composition (Ca2+, ATP, redox status, …) and by various types of regulatory proteins, including kinases and phosphatases as well as by a number of oncogenes and tumor suppressors, but also on their intracellular localization, especially at the ER-mitochondrial and ER-lysosomal interfaces. At these interfaces, Ca2+ microdomains are formed, in which the Ca2+ concentration is finely regulated by the different ER, mitochondrial and lysosomal Ca2+-transport systems and also depends on the functional and structural interactions existing between them. In this review, we therefore discuss the most recent insights in the role of Ca2+ signaling in general, and of the IP3R in particular, in the control of basal mitochondrial bioenergetics, apoptosis, and autophagy at the level of inter-organellar contact sites.
Collapse
Affiliation(s)
- Gemma Roest
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Rita M La Rovere
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| | - Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| |
Collapse
|
85
|
Mirzakhalili E, Epureanu BI, Gourgou E. A mathematical and computational model of the calcium dynamics in Caenorhabditis elegans ASH sensory neuron. PLoS One 2018; 13:e0201302. [PMID: 30048509 PMCID: PMC6062085 DOI: 10.1371/journal.pone.0201302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/28/2018] [Indexed: 12/31/2022] Open
Abstract
We propose a mathematical and computational model that captures the stimulus-generated Ca2+ transients in the C. elegans ASH sensory neuron. The rationale is to develop a tool that will enable a cross-talk between modeling and experiments, using modeling results to guide targeted experimental efforts. The model is built based on biophysical events and molecular cascades known to unfold as part of neurons' Ca2+ homeostasis mechanism, as well as on Ca2+ signaling events. The state of ion channels is described by their probability of being activated or inactivated, and the remaining molecular states are based on biochemically defined kinetic equations or known biochemical motifs. We estimate the parameters of the model using experimental data of hyperosmotic stimulus-evoked Ca2+ transients detected with a FRET sensor in young and aged worms, unstressed and exposed to oxidative stress. We use a hybrid optimization method composed of a multi-objective genetic algorithm and nonlinear least-squares to estimate the model parameters. We first obtain the model parameters for young unstressed worms. Next, we use these values of the parameters as a starting point to identify the model parameters for stressed and aged worms. We show that the model, in combination with experimental data, corroborates literature results. In addition, we demonstrate that our model can be used to predict ASH response to complex combinations of stimulation pulses. The proposed model includes for the first time the ASH Ca2+ dynamics observed during both "on" and "off" responses. This mathematical and computational effort is the first to propose a dynamic model of the Ca2+ transients' mechanism in C. elegans neurons, based on biochemical pathways of the cell's Ca2+ homeostasis machinery. We believe that the proposed model can be used to further elucidate the Ca2+ dynamics of a key C. elegans neuron, to guide future experiments on C. elegans neurobiology, and to pave the way for the development of more mathematical models for neuronal Ca2+ dynamics.
Collapse
Affiliation(s)
- Ehsan Mirzakhalili
- Mechanical Engineering Department, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bogdan I. Epureanu
- Mechanical Engineering Department, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eleni Gourgou
- Mechanical Engineering Department, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, Division of Geriatrics, Medical School, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
86
|
Chen J, Miller M, Unno H, Rosenthal P, Sanderson MJ, Broide DH. Orosomucoid-like 3 (ORMDL3) upregulates airway smooth muscle proliferation, contraction, and Ca 2+ oscillations in asthma. J Allergy Clin Immunol 2018; 142:207-218.e6. [PMID: 28889952 PMCID: PMC5842097 DOI: 10.1016/j.jaci.2017.08.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Airway hyperresponsiveness is a major feature of asthma attributed predominantly to an extrinsic immune/inflammatory response increasing airway smooth muscle (ASM) contractility. OBJECTIVE We investigated whether increased ASM expression of orosomucoid-like 3 (ORMDL3), a gene on chromosome 17q21 highly linked to asthma, induced increased ASM proliferation and contractility in vitro and influenced airway contractility and calcium flux in ASM in precision-cut lung slices (PCLSs) from wild-type and hORMDL3Zp3-Cre mice (which express increased levels of human ORMDL3 [hORMDL3]). METHODS Levels of ASM proliferation and contraction were assessed in ASM cells transfected with ORMDL3 in vitro. In addition, airway contractility and calcium oscillations were quantitated in ASM cells in PCLSs derived from naive wild-type and naive hORMDL3Zp3-Cre mice, which do not have a blood supply. RESULTS Increased ASM expression of ORMDL3 in vitro resulted in increased ASM proliferation and contractility. PCLSs derived from naive hORMDL3Zp3-Cre mice, which do not have airway inflammation, exhibit increased airway contractility with increased calcium oscillations in ASM cells. Increased ASM ORMDL3 expression increases levels of ASM sarcoplasmic reticulum Ca2+ ATPase 2b (SERCA2b), which increases ASM proliferation and contractility. CONCLUSION Overall, these studies provide evidence that an intrinsic increase in ORMDL3 expression in ASM can induce increased ASM proliferation and contractility, which might contribute to increased airway hyperresponsiveness in the absence of airway inflammation in asthmatic patients.
Collapse
Affiliation(s)
- Jun Chen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and the Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Mass
| | - Marina Miller
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Hirotoshi Unno
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Peter Rosenthal
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Michael J Sanderson
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Mass
| | - David H Broide
- Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
87
|
García-Casas P, Arias-Del-Val J, Alvarez-Illera P, Fonteriz RI, Montero M, Alvarez J. Inhibition of Sarco-Endoplasmic Reticulum Ca 2+ ATPase Extends the Lifespan in C. elegans Worms. Front Pharmacol 2018; 9:669. [PMID: 29988547 PMCID: PMC6026643 DOI: 10.3389/fphar.2018.00669] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/04/2018] [Indexed: 12/20/2022] Open
Abstract
The sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) refills the endoplasmic reticulum (ER) with Ca2+ up to the millimolar range and is therefore the main controller of the ER [Ca2+] level ([Ca2+]ER), which has a key role in the modulation of cytosolic Ca2+ signaling and ER-mitochondria Ca2+ transfer. Given that both cytosolic and mitochondrial Ca2+ dynamics strongly interplay with energy metabolism and nutrient-sensitive pathways, both of them involved in the aging process, we have studied the effect of SERCA inhibitors on lifespan in C. elegans. We have used thapsigargin and 2,5-Di-tert-butylhydroquinone (2,5-BHQ) as SERCA inhibitors, and the inactive analog 2,6-Di-tert-butylhydroquinone (2,6-BHQ) as a control for 2,5-BHQ. Every drug was administered to the worms either directly in the agar or via an inclusion compound with γ-cyclodextrin. The results show that 2,6-BHQ produced a small but significant increase in survival, perhaps because of its antioxidant properties. However, 2,5-BHQ produced in all the conditions a much higher increase in lifespan, and the potent and specific SERCA inhibitor thapsigargin also extended the lifespan. The effects of 2,5-BHQ and thapsigargin had a bell-shaped concentration dependence, with a maximum effect at a certain dose and smaller or even toxic effects at higher concentrations. Our data show therefore that submaximal inhibition of SERCA pumps has a pro-longevity effect, suggesting that Ca2+ signaling plays an important role in the aging process and that it could be a promising novel target pathway to act on aging.
Collapse
Affiliation(s)
- Paloma García-Casas
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| | - Jessica Arias-Del-Val
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| | - Pilar Alvarez-Illera
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| | - Rosalba I Fonteriz
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| | - Mayte Montero
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| | - Javier Alvarez
- Department of Biochemistry and Molecular Biology and Physiology, Institute of Biology and Molecular Genetics (IBGM), Faculty of Medicine, University of Valladolid - CSIC, Valladolid, Spain
| |
Collapse
|
88
|
Vervliet T, Robinson EL, Roderick HL. Lnc’ing Ca 2+ , SERCA and cardiac disease. Cell Calcium 2018; 72:132-134. [DOI: 10.1016/j.ceca.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/29/2022]
|
89
|
Yuan L, Wang H, Liu Q, Wang Z, Zhang M, Zhao Y, Liang K, Chen L, Xu T, Xu P. Etoposide-induced protein 2.4 functions as a regulator of the calcium ATPase and protects pancreatic β-cell survival. J Biol Chem 2018; 293:10128-10140. [PMID: 29769309 DOI: 10.1074/jbc.ra118.002399] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/07/2018] [Indexed: 12/22/2022] Open
Abstract
Calcium homeostasis is essential for maintaining the viability and function of pancreatic β cells and plays a key role in preventing the development of diabetes. Decreased levels of ATPase sarcoplasmic/endoplasmic reticulum Ca2+-transporting 2 (ATP2a2), the main calcium pump in β cells, are often found in individuals with diabetes and in diabetic animal models. However, the regulators of ATP2a2 and the molecular mechanisms responsible for controlling ATP2a2 activity remain unclear. Etoposide-induced protein 2.4 (Ei24) is also down-regulated in β cells of diabetic individuals, whereas the effect of decreased Ei24 level on β-cell function is not clarified. Here, using Cre-LoxP and CRISPR/Cas9-based genomic knockout (KO) approaches to generate pancreatic β cell-specific Ei24 KO mice and pancreatic β-cell lines, we found that Ei24 regulates ATP2a2 activity. Specifically, we observed that Ei24 binds to ATP2a2 through Ei24 residues 293-299, which we named here the ATP2a2-interacting region (AIR). Loss of Ei24 inactivated ATP2a2, disrupted calcium homeostasis, and deactivated the calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2)-AMP-activated protein kinase (AMPK) pathway. Elevation of calcium concentration in the endoplasmic reticulum or agonist-induced AMPK activation rescued pancreatic β-cell survival and improved glucose tolerance of Ei24 KO mice. Our findings indicate that targeting the Ei24-ATP2a2 interaction to increase ATP2a2 activity can protect pancreatic β cells and improve glucose homeostasis in diabetic models, suggesting that Ei24 could potentially serve as a target to prevent or manage diabetes.
Collapse
Affiliation(s)
- Lin Yuan
- From the Key Laboratory of RNA Biology and
| | - Huiyu Wang
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | - Qi Liu
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | - Zhe Wang
- From the Key Laboratory of RNA Biology and.,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| | | | - Yan Zhao
- the National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101
| | - Kuo Liang
- the Department of General Surgery, XuanWu Hospital, Capital Medical University, Beijing 100053, and
| | - Liangyi Chen
- the State Key Laboratory of Biomembrane and Membrane Biotechnology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Tao Xu
- the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101, .,the National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101
| | - Pingyong Xu
- From the Key Laboratory of RNA Biology and .,the College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100101
| |
Collapse
|
90
|
Sharma A, Ramena G, Yin Y, Premkumar L, Elble RC. CLCA2 is a positive regulator of store-operated calcium entry and TMEM16A. PLoS One 2018; 13:e0196512. [PMID: 29758025 PMCID: PMC5951673 DOI: 10.1371/journal.pone.0196512] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/13/2018] [Indexed: 11/19/2022] Open
Abstract
The Chloride Channel Accessory (CLCA) protein family was first characterized as regulators of calcium-activated chloride channel (CaCC) currents (ICaCC), but the mechanism has not been fully established. We hypothesized that CLCAs might regulate ICaCC by modulating intracellular calcium levels. In cells stably expressing human CLCA2 or vector, we found by calcium imaging that CLCA2 moderately enhanced intracellular-store release but dramatically increased store-operated entry of calcium upon cytosolic depletion. Moreover, another family member, CLCA1, produced similar effects on intracellular calcium mobilization. Co-immunoprecipitation revealed that CLCA2 interacted with the plasma membrane store-operated calcium channel ORAI-1 and the ER calcium sensor STIM-1. The effect of CLCA2 on ICaCC was tested in HEK293 stably expressing calcium-activated chloride channel TMEM16A. Co-expression of CLCA2 nearly doubled ICaCC in response to a calcium ionophore. These results unveil a new mechanism by which CLCA family members activate ICaCC and suggest a broader role in calcium-dependent processes.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Grace Ramena
- Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Yufang Yin
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Louis Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
| | - Randolph C. Elble
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, United States of America
- * E-mail:
| |
Collapse
|
91
|
van Vliet AR, Sassano ML, Agostinis P. The Unfolded Protein Response and Membrane Contact Sites: Tethering as a Matter of Life and Death? ACTA ACUST UNITED AC 2018. [DOI: 10.1177/2515256418770512] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The endoplasmic reticulum (ER) is the most extensive organelle of the eukaryotic cell and constitutes the major site of protein and lipid synthesis and regulation of intracellular Ca2+ levels. To exert these functions properly, the ER network is shaped in structurally and functionally distinct domains that dynamically remodel in response to intrinsic and extrinsic cues. Moreover, the ER establishes a tight communication with virtually all organelles of the cell through specific subdomains called membrane contact sites. These contact sites allow preferential, nonvesicular channeling of key biological mediators including lipids and Ca2+ between organelles and are harnessed by the ER to interface with and coregulate a variety of organellar functions that are vital to maintain homeostasis. When ER homeostasis is lost, a condition that triggers the activation of an evolutionarily conserved pathway called the unfolded protein response (UPR), the ER undergoes rapid remodeling. These dynamic changes in ER morphology are functionally coupled to the modulation or formation of contact sites with key organelles, such as mitochondria and the plasma membrane, which critically regulate cell fate decisions of the ER-stressed cells. Certain components of the UPR have been shown to facilitate the formation of contact sites through various mechanisms including remodeling of the actin cytoskeleton. In this review, we discuss old and emerging evidence linking the UPR machinery to contact site formation in mammalian cells and discuss their important role in cellular homeostasis.
Collapse
Affiliation(s)
- Alexander R. van Vliet
- Cell Death Research & Therapy Laboratory, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Belgium
| | - Maria Livia Sassano
- Cell Death Research & Therapy Laboratory, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy Laboratory, Department of Cellular and Molecular Medicine, KU Leuven-University of Leuven, Belgium
| |
Collapse
|
92
|
Betzer C, Lassen LB, Olsen A, Kofoed RH, Reimer L, Gregersen E, Zheng J, Calì T, Gai WP, Chen T, Moeller A, Brini M, Fu Y, Halliday G, Brudek T, Aznar S, Pakkenberg B, Andersen JP, Jensen PH. Alpha-synuclein aggregates activate calcium pump SERCA leading to calcium dysregulation. EMBO Rep 2018; 19:embr.201744617. [PMID: 29599149 PMCID: PMC5934765 DOI: 10.15252/embr.201744617] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 02/13/2018] [Accepted: 02/26/2018] [Indexed: 01/03/2023] Open
Abstract
Aggregation of α‐synuclein is a hallmark of Parkinson's disease and dementia with Lewy bodies. We here investigate the relationship between cytosolic Ca2+ and α‐synuclein aggregation. Analyses of cell lines and primary culture models of α‐synuclein cytopathology reveal an early phase with reduced cytosolic Ca2+ levels followed by a later Ca2+ increase. Aggregated but not monomeric α‐synuclein binds to and activates SERCA in vitro, and proximity ligation assays confirm this interaction in cells. The SERCA inhibitor cyclopiazonic acid (CPA) normalises both the initial reduction and the later increase in cytosolic Ca2+. CPA protects the cells against α‐synuclein‐aggregate stress and improves viability in cell models and in Caenorhabditis elegans in vivo. Proximity ligation assays also reveal an increased interaction between α‐synuclein aggregates and SERCA in human brains affected by dementia with Lewy bodies. We conclude that α‐synuclein aggregates bind SERCA and stimulate its activity. Reducing SERCA activity is neuroprotective, indicating that SERCA and down‐stream processes may be therapeutic targets for treating α‐synucleinopathies.
Collapse
Affiliation(s)
- Cristine Betzer
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Louise Berkhoudt Lassen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Olsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Rikke Hahn Kofoed
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lasse Reimer
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emil Gregersen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jin Zheng
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Wei-Ping Gai
- Neuropathological Laboratory, Department of Medicine, Center for Neurological Diseases, University of Adelaide, Adelaide, SA, Australia
| | - Tong Chen
- Department of Medical Biochemistry, School of Medicine, Flinders University, Bedford Park, SA, Australia
| | - Arne Moeller
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Structural Biology, Max Plank Institute of Biophysics, Frankfurt, Germany
| | - Marisa Brini
- Department of Biology, University of Padova, Padova, Italy
| | - Yuhong Fu
- Brain & Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Glenda Halliday
- Brain & Mind Centre, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Tomasz Brudek
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Susana Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Bispebjerg-Frederiksberg Hospital, Copenhagen, Denmark
| | | | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark .,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
93
|
Vervliet T. Ryanodine Receptors in Autophagy: Implications for Neurodegenerative Diseases? Front Cell Neurosci 2018; 12:89. [PMID: 29636667 PMCID: PMC5880912 DOI: 10.3389/fncel.2018.00089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 12/18/2022] Open
Abstract
Intracellular Ca2+ signaling is important in the regulation of several cellular processes including autophagy. The endoplasmic reticulum (ER) is the main and largest intracellular Ca2+ store. At the ER two protein families of Ca2+ release channels, inositol 1,4,5-trisphosphate receptors (IP3Rs) and ryanodine receptors (RyRs), are expressed. Several studies have reported roles in the regulation of autophagy for the ubiquitously expressed IP3R. For instance, IP3R-mediated Ca2+ release supresses basal autophagic flux by promoting mitochondrial metabolism, while also promoting the rapid initial increase in autophagic flux in response to nutrient starvation. Insights into the contribution of RyRs in autophagy have been lagging significantly compared to the advances made for IP3Rs. This is rather surprising considering that RyRs are predominantly expressed in long-lived cells with specialized metabolic needs, such as neurons and muscle cells, in which autophagy plays important roles. In this review article, recent studies revealing roles for RyRs in the regulation of autophagy will be discussed. Several RyR-interacting proteins that have been established to modulate both RyR function and autophagy will also be highlighted. Finally, the involvement of RyRs in neurodegenerative diseases will be addressed. Inhibition of RyR channels has not only been shown to be beneficial for treating several of these diseases but also regulates autophagy.
Collapse
Affiliation(s)
- Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
94
|
Calcium Dynamics as a Machine for Decoding Signals. Trends Cell Biol 2018; 28:258-273. [PMID: 29409699 DOI: 10.1016/j.tcb.2018.01.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 11/22/2022]
Abstract
Calcium (Ca2+) is considered one of the most-important biological cations, because it is implicated in cell physiopathology and cell fate through a finely tuned signaling system. In support of this notion, Ca2+ is the primary driver of cell proliferation and cell growth; however, it is also intimately linked to cell death. Functional abnormalities or mutations in proteins that mediate Ca2+ homeostasis usually lead to a plethora of diseases and pathogenic states, including cancer, heart failure, diabetes, and neurodegenerative disease. In this review, we examine recent discoveries in the highly localized nature of Ca2+-dependent signal transduction and its roles in cell fate, inflammasome activation, and synaptic transmission.
Collapse
|
95
|
Mikkelsen SA, Vangheluwe P, Andersen JP. A Darier disease mutation relieves kinetic constraints imposed by the tail of sarco(endo)plasmic reticulum Ca 2+-ATPase 2b. J Biol Chem 2018; 293:3880-3889. [PMID: 29363575 DOI: 10.1074/jbc.ra117.000941] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/19/2018] [Indexed: 11/06/2022] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) 2b isoform possesses an extended C terminus (SERCA2b tail) forming an 11th transmembrane (TM) helix, which slows conformational changes of the Ca2+-pump reaction cycle. Here, we report that a Darier disease (DD) mutation of SERCA2b that changes a glutamate to a lysine in the cytoplasmic loop between TM8 and TM9 (E917K) relieves these kinetic constraints. We analyzed the effects of this mutation on the overall reaction and the individual partial reactions of the Ca2+ pump compared with the corresponding mutations of the SERCA2a and SERCA1a isoforms, lacking the SERCA2b tail. In addition to a reduced affinity for Ca2+, caused by the mutation in all three isoforms examined, we observed a unique enhancing effect on the turnover rates of ATPase activity and Ca2+ transport for the SERCA2b E917K mutation. This relief of kinetic constraints contrasted with inhibitory effects observed for the corresponding SERCA2a and SERCA1a (E918K) mutations. These observations indicated that the E917K/E918K mutations affect the rate-limiting conformational change in isoform-specific ways and that the SERCA2b mutation perturbs the interactions of TM11 with other SERCA2b regions. Mutational analysis of an arginine in TM7 that interacts with the glutamate in SERCA1a crystal structures suggested that in wildtype SERCA2b, the corresponding arginine (Arg-835) may be involved in mediating the conformational restriction by TM11. Moreover, the E917K mutation may disturb TM11 through the cytoplasmic loop between TM10 and TM11. In conclusion, our findings have identified structural elements of importance for the kinetic constraints imposed by TM11.
Collapse
Affiliation(s)
- Stine A Mikkelsen
- From the Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark and
| | - Peter Vangheluwe
- the Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium
| | - Jens Peter Andersen
- From the Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark and
| |
Collapse
|
96
|
Sousa L, Pessoa MTC, Costa TGF, Cortes VF, Santos HL, Barbosa LA. Iron overload impact on P-ATPases. Ann Hematol 2018; 97:377-385. [PMID: 29307086 DOI: 10.1007/s00277-017-3222-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 12/23/2017] [Indexed: 12/16/2022]
Abstract
Iron is a chemical element that is active in the fundamental physiological processes for human life, but its burden can be toxic to the body, mainly because of the stimulation of membrane lipid peroxidation. For this reason, the action of iron on many ATPases has been studied, especially on P-ATPases, such as the Na+,K+-ATPase and the Ca2+-ATPase. On the Fe2+-ATPase activity, the free iron acts as an activator, decreasing the intracellular Fe2+ and playing a protection role for the cell. On the Ca2+-ATPase activity, the iron overload decreases the enzyme activity, raising the cytoplasmic Ca2+ and decreasing the sarco/endoplasmic reticulum and the Golgi apparatus Ca2+ concentrations, which could promote an enzyme oxidation, nitration, and fragmentation. However, the iron overload effect on the Na+,K+-ATPase may change according to the tissue expressions. On the renal cells, as well as on the brain and the heart, iron promotes an enzyme inactivation, whereas its effect on the erythrocytes seems to be the opposite, directly stimulating the ATPase activity, or stimulating it by signaling pathways involving ROS and PKC. Modulations in the ATPase activity may impair the ionic transportation, which is essential for cell viability maintenance, inducing irreversible damage to the cell homeostasis. Here, we will discuss about the iron overload effect on the P-ATPases, such as the Na+,K+-ATPase, the Ca2+-ATPase, and the Fe2+-ATPase.
Collapse
Affiliation(s)
- Leilismara Sousa
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil
| | - Marco Tulio C Pessoa
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil
| | - Tamara G F Costa
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil
| | - Vanessa F Cortes
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil
| | - Herica L Santos
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del Rei, Av Sebastião Gonçalves Coelho, 400, Divinópolis, MG, 35501-296, Brazil.
| |
Collapse
|
97
|
Wasąg P, Suwińska A, Zakrzewski P, Walczewski J, Lenartowski R, Lenartowska M. Calreticulin localizes to plant intra/extracellular peripheries of highly specialized cells involved in pollen-pistil interactions. PROTOPLASMA 2018; 255:57-67. [PMID: 28620697 PMCID: PMC5756280 DOI: 10.1007/s00709-017-1134-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/05/2017] [Indexed: 05/11/2023]
Abstract
Calcium (Ca2+) plays essential roles in generative reproduction of angiosperms, but the sites and mechanisms of Ca2+ storage and mobilization during pollen-pistil interactions have not been fully defined. Both external and internal Ca2+ stores are likely important during male gametophyte communication with the sporophytic and gametophytic cells within the pistil. Given that calreticulin (CRT), a Ca2+-buffering protein, is able to bind Ca2+ reversibly, it can serve as a mobile store of easily releasable Ca2+ (so called an exchangeable Ca2+) in eukaryotic cells. CRT has typical endoplasmic reticulum (ER) targeting and retention signals and resides primarily in the ER. However, localization of this protein outside the ER has also been revealed in both animal and plant cells, including Golgi/dictyosomes, nucleus, plasma membrane/cell surface, plasmodesmata, and even extracellular matrix. These findings indicate that CRT may function in a variety of different cell compartments and specialized structures. We have recently shown that CRT is highly expressed and accumulated in the ER of plant cells involved in pollen-pistil interactions in Petunia, and we proposed an essential role for CRT in intracellular Ca2+ storage and mobilization during the key reproductive events. Here, we demonstrate that both CRT and exchangeable Ca2+ are localized in the intra/extracellular peripheries of highly specialized plant cells, such as the pistil transmitting tract cells, pollen tubes, nucellus cells surrounding the embryo sac, and synergids. Based on our present results, we propose that extracellularly located CRT is also involved in Ca2+ storage and mobilization during sexual reproduction of angiosperms.
Collapse
Affiliation(s)
- Piotr Wasąg
- Laboratory of Isotope and Instrumental Analysis, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Anna Suwińska
- Laboratory of Developmental Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Przemysław Zakrzewski
- Laboratory of Developmental Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Jakub Walczewski
- Department of Plant Pathology, Plant Breeding and Acclimatization Institute, National Research Institute, Radzików, Poland
| | - Robert Lenartowski
- Laboratory of Isotope and Instrumental Analysis, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Marta Lenartowska
- Laboratory of Developmental Biology, Faculty of Biology and Environmental Protection, Nicolaus Copernicus University in Toruń, Toruń, Poland.
| |
Collapse
|
98
|
Luarte A, Cornejo VH, Bertin F, Gallardo J, Couve A. The axonal endoplasmic reticulum: One organelle-many functions in development, maintenance, and plasticity. Dev Neurobiol 2017; 78:181-208. [PMID: 29134778 DOI: 10.1002/dneu.22560] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 11/02/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is highly conserved in eukaryotes and neurons. Indeed, the localization of the organelle in axons has been known for nearly half a century. However, the relevance of the axonal ER is only beginning to emerge. In this review, we discuss the structure of the ER in axons, examining the role of ER-shaping proteins and highlighting reticulons. We analyze the multiple functions of the ER and their potential contribution to axonal physiology. First, we examine the emerging roles of the axonal ER in lipid synthesis, protein translation, processing, quality control, and secretory trafficking of transmembrane proteins. We also review the impact of the ER on calcium dynamics, focusing on intracellular mechanisms and functions. We describe the interactions between the ER and endosomes, mitochondria, and synaptic vesicles. Finally, we analyze available proteomic data of axonal preparations to reveal the dynamic functionality of the ER in axons during development. We suggest that the dynamic proteome and a validated axonal interactome, together with state-of-the-art methodologies, may provide interesting research avenues in axon physiology that may extend to pathology and regeneration. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 181-208, 2018.
Collapse
Affiliation(s)
- Alejandro Luarte
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Víctor Hugo Cornejo
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisca Bertin
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Javiera Gallardo
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrés Couve
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
99
|
Yoboue ED, Rimessi A, Anelli T, Pinton P, Sitia R. Regulation of Calcium Fluxes by GPX8, a Type-II Transmembrane Peroxidase Enriched at the Mitochondria-Associated Endoplasmic Reticulum Membrane. Antioxid Redox Signal 2017; 27:583-595. [PMID: 28129698 DOI: 10.1089/ars.2016.6866] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
UNLABELLED Glutathione peroxidases (GPXs) are enzymes that are present in almost all organisms with the primary function of limiting peroxide accumulation. In mammals, two of the eight members (GPX7 and GPX8) reside in the endoplasmic reticulum (ER). A peculiar feature of GPX8 is the concomitant presence of a conserved N-terminal transmembrane domain (TMD) and a C-terminal KDEL-like motif for ER localization. AIMS Investigating whether and how GPX8 impacts Ca2+ homeostasis and signaling. RESULTS We show that GPX8 is enriched in mitochondria-associated membranes and regulates Ca2+ storage and fluxes. Its levels correlate with [Ca2+]ER, and cytosolic and mitochondrial Ca2+ fluxes. GPX7, which lacks a TMD, does not share these properties. Deleting or replacing the GPX8 TMD with an unrelated N-terminal membrane integration sequence abolishes all effects on Ca2+ fluxes, whereas appending the GPX8 TMD to GPX7 transfers the Ca2+-regulating properties. Innovation and Conclusion: The notion that the TMD of GPX8, in addition to its enzymatic activity, is essential for regulating Ca2+ dynamics reveals a novel level of integration between redox-related proteins and Ca2+ signaling/homeostasis. Antioxid. Redox Signal. 27, 583-595.
Collapse
Affiliation(s)
- Edgar Djaha Yoboue
- 1 Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele , Milan, Italy
| | - Alessandro Rimessi
- 2 Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara, Italy
| | - Tiziana Anelli
- 1 Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele , Milan, Italy .,3 Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Paolo Pinton
- 2 Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), Department of Morphology, Surgery and Experimental Medicine, University of Ferrara , Ferrara, Italy
| | - Roberto Sitia
- 1 Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele , Milan, Italy .,3 Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| |
Collapse
|
100
|
The ER-Localized Transmembrane Protein EPG-3/VMP1 Regulates SERCA Activity to Control ER-Isolation Membrane Contacts for Autophagosome Formation. Mol Cell 2017; 67:974-989.e6. [PMID: 28890335 DOI: 10.1016/j.molcel.2017.08.005] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/23/2017] [Accepted: 08/08/2017] [Indexed: 01/27/2023]
Abstract
During autophagosome formation in mammalian cells, isolation membranes (IMs; autophagosome precursors) dynamically contact the ER. Here, we demonstrated that the ER-localized metazoan-specific autophagy protein EPG-3/VMP1 controls ER-IM contacts. Loss of VMP1 causes stable association of IMs with the ER, thus blocking autophagosome formation. Interaction of WIPI2 with the ULK1/FIP200 complex and PI(3)P contributes to the formation of ER-IM contacts, and these interactions are enhanced by VMP1 depletion. VMP1 controls contact formation by promoting SERCA (sarco[endo]plasmic reticulum calcium ATPase) activity. VMP1 interacts with SERCA and prevents formation of the SERCA/PLN/SLN inhibitory complex. VMP1 also modulates ER contacts with lipid droplets, mitochondria, and endosomes. These ER contacts are greatly elevated by the SERCA inhibitor thapsigargin. Calmodulin acts as a sensor/effector to modulate the ER contacts mediated by VMP1/SERCA. Our study provides mechanistic insights into the establishment and disassociation of ER-IM contacts and reveals that VMP1 modulates SERCA activity to control ER contacts.
Collapse
|