1
|
Rencber SF, Yazır Y, Sarıhan M, Sezer Z, Korun ZEU, Ozturk A, Duruksu G, Guzel E, Akpınar G, Corakci A. Endoplasmic reticulum stress of endometrial mesenchymal stem cells in endometriosis. Tissue Cell 2024; 91:102544. [PMID: 39217786 DOI: 10.1016/j.tice.2024.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE The human endometrium has significant regenerative abilities due to stem cells, which are vital in immunomodulation, immune tolerance, steroid hormone response, and inflammation. Endometriosis, an inflammatory gynecological disorder where endometrium-like tissue grows outside uterus, affects millions of women and often causes infertility. Recent research indicates that stem cells contribute to pathology of endometriosis. ER stress is implicated in various diseases, including endometriosis. This study aims to examine ER stress in eMSCs within endometriosis pathogenesis and uncover underlying disease mechanisms. METHODS Samples were collected from healthy subjects and women with endometriosis in both proliferative and secretory phases. eMSCs were isolated and characterized via flow cytometry. ER stress protein levels were assessed using proteomic analysis, with validation through Western Blot and immunofluorescence staining. Gene expression was analyzed by RT-qPCR, and ultrastructural examination of eMSCs was conducted using TEM. ER stress markers in tissue samples were detected in SUSD2+ eMSCs through immunofluorescence staining and visualized using a confocal microscope. Statistical analysis was performed using SPSS program. RESULTS The proteomics analysis uncovered ER stress-related proteins (DDRGK1, RTN3, ERp44, TMED2, TMEM33, TMX3) whose levels were significantly distinct from control group. Western Blot analysis and immunofluorescence staining results at protein level; RT-qPCR results at gene level supported these findings. TEM analysis also showed ultrastructural presence of ER stress in endometriosis groups. CONCLUSION Presence of ER stress in eMSCs in pathogenesis of endometriosis has been demonstrated using various methods. Our research has potential to shed light on pathology of endometriosis and offer promising avenues for non-invasive diagnosis and potential treatment.
Collapse
Affiliation(s)
- Selenay Furat Rencber
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Yusufhan Yazır
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey.
| | - Mehmet Sarıhan
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Zehra Sezer
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeynep Ece Utkan Korun
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University, İstanbul, Turkey
| | - Ahmet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey; Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Gokhan Duruksu
- Department of Stem Cell, Institute of Health Sciences, Kocaeli University, Kocaeli, Turkey; Center for Stem Cell and Gene Therapies Research and Practice, Kocaeli University, Kocaeli, Turkey
| | - Elif Guzel
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gurler Akpınar
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Aydın Corakci
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
2
|
Huttner IG, Santiago CF, Jacoby A, Cheng D, Trivedi G, Cull S, Cvetkovska J, Chand R, Berger J, Currie PD, Smith KA, Fatkin D. Loss of Sec-1 Family Domain-Containing 1 ( scfd1) Causes Severe Cardiac Defects and Endoplasmic Reticulum Stress in Zebrafish. J Cardiovasc Dev Dis 2023; 10:408. [PMID: 37887855 PMCID: PMC10607167 DOI: 10.3390/jcdd10100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder that frequently leads to heart failure, arrhythmias, and death. While DCM is often heritable, disease-causing mutations are identified in only ~30% of cases. In a forward genetic mutagenesis screen, we identified a novel zebrafish mutant, heart and head (hahvcc43), characterized by early-onset cardiomyopathy and craniofacial defects. Linkage analysis and next-generation sequencing identified a nonsense variant in the highly conserved scfd1 gene, also known as sly1, that encodes sec1 family domain-containing 1. Sec1/Munc18 proteins, such as Scfd1, are involved in membrane fusion regulating endoplasmic reticulum (ER)/Golgi transport. CRISPR/Cas9-engineered scfd1vcc44 null mutants showed severe cardiac and craniofacial defects and embryonic lethality that recapitulated the phenotype of hahvcc43 mutants. Electron micrographs of scfd1-depleted cardiomyocytes showed reduced myofibril width and sarcomere density, as well as reticular network disorganization and fragmentation of Golgi stacks. Furthermore, quantitative PCR analysis showed upregulation of ER stress response and apoptosis markers. Both heterozygous hahvcc43 mutants and scfd1vcc44 mutants survived to adulthood, showing chamber dilation and reduced ventricular contraction. Collectively, our data implicate scfd1 loss-of-function as the genetic defect at the hahvcc43 locus and provide new insights into the role of scfd1 in cardiac development and function.
Collapse
Affiliation(s)
- Inken G. Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Celine F. Santiago
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Arie Jacoby
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Delfine Cheng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Gunjan Trivedi
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Stephen Cull
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Jasmina Cvetkovska
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Renee Chand
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Kelly A. Smith
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
- Cardiology Department, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
3
|
Jeon HJ, Cho Y, Kim K, Kim C, Lee SE. Combined toxicity of 3,5,6-trichloro-2-pyridinol and 2-(bromomethyl)naphthalene in the early stages of zebrafish (Danio rerio) embryos: Abnormal heart development at lower concentrations via differential expression of heart forming-related genes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 325:121450. [PMID: 36940914 DOI: 10.1016/j.envpol.2023.121450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
Combined toxicity can occur in the environment according to the combination of single substances, and the combination works additively or in a synergistic or antagonistic mode. In our study, 3,5,6-trichloro-2-pyridinol (TCP) and 2-(bromomethyl)naphthalene (2-BMN) were used to measure combined toxicity in zebrafish (Danio rerio) embryos. As the lethal concentration (LC) values were obtained through single toxicity, the lethal effects at all combinational concentrations were considered synergistic by the Independent Action model. At 96 hpf, the combined toxicity of TCP LC10 + 2-BMN LC10, the lowest combinational concentration, resulted in high mortality, strong inhibition of hatching, and various morphological changes in zebrafish embryos. Combined treatment resulted in the downregulation of cyp1a, leading to reduced detoxification of the treated chemicals in embryos. These combinations may enhance endocrine-disrupting properties via upregulation of vtg1 in embryos, and inflammatory responses and endoplasmic reticulum stress were found to upregulate il-β, atf4, and atf6. These combinations might induce severe abnormal cardiac development in embryos via downregulation of myl7, cacna1c, edn1, and vmhc expression, and upregulation of the nppa gene. Therefore, the combined toxicity of these two chemicals was observed in zebrafish embryos, which proves that similar substances can exhibit stronger combined toxicity than single toxicity.
Collapse
Affiliation(s)
- Hwang-Ju Jeon
- Red River Research Station, Louisiana State University Agricultural Center, Bossier City, LA, USA
| | - Yerin Cho
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyeongnam Kim
- Institute of Quality and Safety Evaluation of Agricultural Products, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Chaeeun Kim
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sung-Eun Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea; Institute of Quality and Safety Evaluation of Agricultural Products, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Integrative Biology, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
4
|
Amagai Y, Yamada M, Kowada T, Watanabe T, Du Y, Liu R, Naramoto S, Watanabe S, Kyozuka J, Anelli T, Tempio T, Sitia R, Mizukami S, Inaba K. Zinc homeostasis governed by Golgi-resident ZnT family members regulates ERp44-mediated proteostasis at the ER-Golgi interface. Nat Commun 2023; 14:2683. [PMID: 37160917 PMCID: PMC10170084 DOI: 10.1038/s41467-023-38397-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/01/2023] [Indexed: 05/11/2023] Open
Abstract
Many secretory enzymes acquire essential zinc ions (Zn2+) in the Golgi complex. ERp44, a chaperone operating in the early secretory pathway, also binds Zn2+ to regulate its client binding and release for the control of protein traffic and homeostasis. Notably, three membrane transporter complexes, ZnT4, ZnT5/ZnT6 and ZnT7, import Zn2+ into the Golgi lumen in exchange with protons. To identify their specific roles, we here perform quantitative Zn2+ imaging using super-resolution microscopy and Zn2+-probes targeted in specific Golgi subregions. Systematic ZnT-knockdowns reveal that ZnT4, ZnT5/ZnT6 and ZnT7 regulate labile Zn2+ concentration at the distal, medial, and proximal Golgi, respectively, consistent with their localization. Time-course imaging of cells undergoing synchronized secretory protein traffic and functional assays demonstrates that ZnT-mediated Zn2+ fluxes tune the localization, trafficking, and client-retrieval activity of ERp44. Altogether, this study provides deep mechanistic insights into how ZnTs control Zn2+ homeostasis and ERp44-mediated proteostasis along the early secretory pathway.
Collapse
Affiliation(s)
- Yuta Amagai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Momo Yamada
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Toshiyuki Kowada
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Tomomi Watanabe
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Yuyin Du
- Department of Chemistry, Faculty of Science, Tohoku University, 6-3 Aramaki-aza-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Rong Liu
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Satoshi Naramoto
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Kita 10 Nishi 8, Kita-ku, Sapporo, Japan
| | - Satoshi Watanabe
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Junko Kyozuka
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
| | - Tiziana Anelli
- Division of Genetics and Cell Biology, Vita-Salute University, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Tiziana Tempio
- Division of Genetics and Cell Biology, Vita-Salute University, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Vita-Salute University, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Shin Mizukami
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda, Tokyo, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, Miyagi, 980-8578, Japan.
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan.
- Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda, Tokyo, Japan.
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
5
|
Hamilton S, Terentyev D. ER stress and calcium-dependent arrhythmias. Front Physiol 2022; 13:1041940. [PMID: 36425292 PMCID: PMC9679650 DOI: 10.3389/fphys.2022.1041940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
The sarcoplasmic reticulum (SR) plays the key role in cardiac function as the major source of Ca2+ that activates cardiomyocyte contractile machinery. Disturbances in finely-tuned SR Ca2+ release by SR Ca2+ channel ryanodine receptor (RyR2) and SR Ca2+ reuptake by SR Ca2+-ATPase (SERCa2a) not only impair contraction, but also contribute to cardiac arrhythmia trigger and reentry. Besides being the main Ca2+ storage organelle, SR in cardiomyocytes performs all the functions of endoplasmic reticulum (ER) in other cell types including protein synthesis, folding and degradation. In recent years ER stress has become recognized as an important contributing factor in many cardiac pathologies, including deadly ventricular arrhythmias. This brief review will therefore focus on ER stress mechanisms in the heart and how these changes can lead to pro-arrhythmic defects in SR Ca2+ handling machinery.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States,*Correspondence: Shanna Hamilton,
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
6
|
Tian Y, Sun H, Bao Y, Feng H, Pang J, En R, Jiang H, Wang T. ERp44 Regulates the Proliferation, Migration, Invasion, and Apoptosis of Gastric Cancer Cells Via Activation of ER Stress. Biochem Genet 2022; 61:809-822. [PMID: 36178559 DOI: 10.1007/s10528-022-10281-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 09/01/2022] [Indexed: 11/24/2022]
Abstract
Gastric cancer (GC) is one of the most prevalent malignancies worldwide. Endoplasmic reticulum (ER) stress plays a key role in the progression of GC. Rapid proliferation of tumor cells interferes with ER homeostasis, leading to ER stress and triggering unfolded protein response. Therefore, it is very necessary to investigate abnormally expressed ER resident proteins (ERp) in cancer cells. This study aimed to investigate the possible roles of ERp44. The mRNA and protein expression of genes were detected using qRT-PCR and western blot. Cell apoptosis was calculated using flow cytometry. Cell proliferation was determined using CCK-8 and colony formation assay. Cell migration was detected by wound healing, and cell invasion was measured by transwell assay. We found that ERp44 was obviously decreased in GC tissues. Furthermore, ERp44 overexpression distinctly suppressed the proliferation, migration, and invasion of MGC-803 and KATO III cells. In contrast, apoptosis was promoted by ERp44 overexpression. Furthermore, mechanistic studies revealed that overexpression of ERp44 inhibited malignant biological processes by regulating the eIF-2α/CHOP signaling pathway. Taken together, our data demonstrated that ERp44 regulated the proliferation, migration, invasion, and apoptosis via ERp44/eIF-2α/CHOP axis in GC. Targeting the ERp44and ER stress may be a promising strategy for GC.
Collapse
Affiliation(s)
- Yongjing Tian
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Haibin Sun
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Yinshengboer Bao
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Haiping Feng
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Jian Pang
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Riletu En
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Hongliang Jiang
- Department of Gastrointestinal Surgery, Inner Mongolia Bayannur Hospital, Inner Mongolia, 015000, China
| | - Tengqi Wang
- Department of Cancer Center, Inner Mongolia Bayannur Hospital, No. 98, Ulan Buhe Road, Bayan Nur, Inner Mongolia, 015000, China.
| |
Collapse
|
7
|
Carreras-Sureda A, Kroemer G, Cardenas JC, Hetz C. Balancing energy and protein homeostasis at ER-mitochondria contact sites. Sci Signal 2022; 15:eabm7524. [DOI: 10.1126/scisignal.abm7524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell and participates in multiple essential functions, including the production of secretory proteins, lipid synthesis, and calcium storage. Sustaining proteostasis requires an intimate coupling with energy production. Mitochondrial respiration evolved to be functionally connected to ER physiology through a physical interface between both organelles known as mitochondria-associated membranes. This quasi-synaptic structure acts as a signaling hub that tunes the function of both organelles in a bidirectional manner and controls proteostasis, cell death pathways, and mitochondrial bioenergetics. Here, we discuss the main signaling mechanisms governing interorganellar communication and their putative role in diseases including cancer and neurodegeneration.
Collapse
Affiliation(s)
- Amado Carreras-Sureda
- Department of Cell Physiology and Metabolism, University of Geneva, 1, rue Michel-Servet, 1211 Geneva, Switzerland
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Julio Cesar Cardenas
- Center for Integrative Biology, Mayor University, 7510041 Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, 70086 Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Claudio Hetz
- Center for Geroscience, Brain Health, and Metabolism, 70086 Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, 70086 Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, 70086 Santiago, Chile
| |
Collapse
|
8
|
Hamilton S, Terentyeva R, Bogdanov V, Kim TY, Perger F, Yan J, Ai X, Carnes CA, Belevych AE, George CH, Davis JP, Gyorke S, Choi BR, Terentyev D. Ero1α-Dependent ERp44 Dissociation From RyR2 Contributes to Cardiac Arrhythmia. Circ Res 2022; 130:711-724. [PMID: 35086342 PMCID: PMC8893133 DOI: 10.1161/circresaha.121.320531] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Oxidative stress in cardiac disease promotes proarrhythmic disturbances in Ca2+ homeostasis, impairing luminal Ca2+ regulation of the sarcoplasmic reticulum (SR) Ca2+ release channel, the RyR2 (ryanodine receptor), and increasing channel activity. However, exact mechanisms underlying redox-mediated increase of RyR2 function in cardiac disease remain elusive. We tested whether the oxidoreductase family of proteins that dynamically regulate the oxidative environment within the SR are involved in this process. METHODS A rat model of hypertrophy induced by thoracic aortic banding (TAB) was used for ex vivo whole heart optical mapping and for Ca2+ and reactive oxygen species imaging in isolated ventricular myocytes (VMs). RESULTS The SR-targeted reactive oxygen species biosensor ERroGFP showed increased intra-SR oxidation in TAB VMs that was associated with increased expression of Ero1α (endoplasmic reticulum oxidoreductase 1 alpha). Pharmacological (EN460) or genetic Ero1α inhibition normalized SR redox state, increased Ca2+ transient amplitude and SR Ca2+ content, and reduced proarrhythmic spontaneous Ca2+ waves in TAB VMs under β-adrenergic stimulation (isoproterenol). Ero1α overexpression in Sham VMs had opposite effects. Ero1α inhibition attenuated Ca2+-dependent ventricular tachyarrhythmias in TAB hearts challenged with isoproterenol. Experiments in TAB VMs and human embryonic kidney 293 cells expressing human RyR2 revealed that an Ero1α-mediated increase in SR Ca2+-channel activity involves dissociation of intraluminal protein ERp44 (endoplasmic reticulum protein 44) from the RyR2 complex. Site-directed mutagenesis and molecular dynamics simulations demonstrated a novel redox-sensitive association of ERp44 with RyR2 mediated by intraluminal cysteine 4806. ERp44-RyR2 association in TAB VMs was restored by Ero1α inhibition, but not by reducing agent dithiothreitol, as hypo-oxidation precludes formation of covalent bond between RyR2 and ERp44. CONCLUSIONS A novel axis of intraluminal interaction between RyR2, ERp44, and Ero1α has been identified. Ero1α inhibition exhibits promising therapeutic potential by stabilizing RyR2-ERp44 complex, thereby reducing spontaneous Ca2+ release and Ca2+-dependent tachyarrhythmias in hypertrophic hearts, without causing hypo-oxidative stress in the SR.
Collapse
Affiliation(s)
- Shanna Hamilton
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Tae Yun Kim
- Cardiovascular Research Center, Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (T.Y.K., B.-R.C.)
| | - Fruzsina Perger
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Jiajie Yan
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Xun Ai
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Cynthia A. Carnes
- College of Pharmacy (C.A.C.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Andriy E. Belevych
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | | | - Jonathan P. Davis
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Sandor Gyorke
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| | - Bum-Rak Choi
- Cardiovascular Research Center, Rhode Island Hospital, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI (T.Y.K., B.-R.C.)
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology (S.H., R.T., V.B., F.P., J.Y., X.A., A.E.B., J.P.D., S.G., D.T.), The Ohio State University.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus (S.H., R.T., V.B., F.P., J.Y., X.A., C.A.C., A.E.B., J.P.D., S.G., D.T.)
| |
Collapse
|
9
|
Bi Y, Yang Z, Jin M, Zhai K, Wang J, Mao Y, Liu Y, Ding M, Wang H, Wang F, Cai H, Ji G. ERp44 is required for endocardial cushion development by regulating VEGFA secretion in myocardium. Cell Prolif 2022; 55:e13179. [PMID: 35088919 PMCID: PMC8891561 DOI: 10.1111/cpr.13179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Endocardial cushions are precursors of the valve septum complex that separates the four heart chambers. Several genes have been implicated in the development of endocardial cushions. Specifically, ERp44 has been found to play a role in the early secretory pathway, but its function in heart development has not been well studied. MATERIALS AND METHODS In this study, we established conditional and tissue-specific knockout mouse models. The morphology, survival rate, the development of heart and endocardial cushion were under evaluation. The relationship between ERp44 and VEGFA was investigated by transcriptome, qPCR, WB, immunofluorescence and immunohistochemistry. RESULTS ERp44 knockout (KO) mice were smaller in size, and most mice died during early postnatal life. KO hearts exhibited the typical phenotypes of congenital heart diseases, such as abnormal heart shapes and severe septal and valvular defects. Similar phenotypes were found in cTNT-Cre+/- ; ERp44fl / fl mice, which indicated that myocardial ERp44 principally controls endocardial cushion formation. Further studies demonstrated that the deletion of ERp44 significantly decreased the proliferation of cushion cells and impaired the endocardial-mesenchymal transition (EndMT), which was followed by endocardial cushion dysplasia. Finally, we found that ERp44 was directly bound to VEGFA and controlled its release, further regulating EndMT. CONCLUSION We demonstrated that ERp44 plays a specific role in heart development. ERp44 contributes to the development of the endocardial cushion by affecting VEGFA-mediated EndMT.
Collapse
Affiliation(s)
- Youkun Bi
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhiguang Yang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Meng Jin
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Kui Zhai
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jun Wang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Mao
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Liu
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Mingqin Ding
- National Institute of Biological SciencesBeijingChina
| | - Huiwen Wang
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Fengchao Wang
- National Institute of Biological SciencesBeijingChina
| | - Hong Cai
- Department of DermatologyAir Force Medical CenterPLABeijingChina
| | - Guangju Ji
- Key Laboratory of Interdisciplinary ResearchInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| |
Collapse
|
10
|
Bi Y, Chang Y, Liu Q, Mao Y, Zhai K, Zhou Y, Jiao R, Ji G. ERp44/CG9911 promotes fat storage in Drosophila adipocytes by regulating ER Ca 2+ homeostasis. Aging (Albany NY) 2021; 13:15013-15031. [PMID: 34031268 PMCID: PMC8221293 DOI: 10.18632/aging.203063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/27/2021] [Indexed: 12/12/2022]
Abstract
Fat storage is one of the important strategies employed in regulating energy homeostasis. Impaired lipid storage causes metabolic disorders in both mammals and Drosophila. In this study, we report CG9911, the Drosophila homolog of ERp44 (endoplasmic reticulum protein 44) plays a role in regulating adipose tissue fat storage. Using the CRISPR/Cas9 system, we generated a CG9911 mutant line deleting 5 bp of the coding sequence. The mutant flies exhibit phenotypes of lower bodyweight, fewer lipid droplets, reduced TAG level and increased expression of lipolysis related genes. The increased lipolysis phenotype is enhanced in the presence of ER stresses and suppressed by a reduction of the ER Ca2+. Moreover, loss of CG9911 per se results in a decrease of ER Ca2+ in the fat body. Together, our results reveal a novel function of CG9911 in promoting fat storage via regulating ER Ca2+ signal in Drosophila.
Collapse
Affiliation(s)
- Youkun Bi
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Chang
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qun Liu
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Mao
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Zhai
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Zhou
- Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Renjie Jiao
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China.,Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 510182, China
| | - Guangju Ji
- Key Laboratory of Interdisciplinary Research, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
11
|
Mo G, Liu X, Zhong Y, Mo J, Li Z, Li D, Zhang L, Liu Y. IP3R1 regulates Ca 2+ transport and pyroptosis through the NLRP3/Caspase-1 pathway in myocardial ischemia/reperfusion injury. Cell Death Dis 2021; 7:31. [PMID: 33568649 PMCID: PMC7876122 DOI: 10.1038/s41420-021-00404-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 01/31/2023]
Abstract
Intracellular ion channel inositol 1,4,5-triphosphate receptor (IP3R1) releases Ca2+ from endoplasmic reticulum. The disturbance of IP3R1 is related to several neurodegenerative diseases. This study investigated the mechanism of IP3R1 in myocardial ischemia/reperfusion (MI/R). After MI/R modeling, IP3R1 expression was silenced in myocardium of MI/R rats to explore its role in the concentration of myocardial enzymes, infarct area, Ca2+ level, NLRP3/Caspase-1, and pyroptosis markers and inflammatory factors. The adult rat cardiomyocytes were isolated and cultured to establish hypoxia/reperfusion (H/R) cell model. The expression of IP3R1 was downregulated or ERP44 was overexpressed in H/R-induced cells. Nifedipine D6 was added to H/R-induced cells to block Ca2+ channel or Nigericin was added to activate NLRP3. IP3R1 was highly expressed in myocardium of MI/R rats, and silencing IP3R1 alleviated MI/R injury, reduced Ca2+ overload, inflammation and pyroptosis in MI/R rats, and H/R-induced cells. The binding of ERP44 to IP3R1 inhibited Ca2+ overload, alleviated cardiomyocyte inflammation, and pyroptosis. The increase of intracellular Ca2+ level caused H/R-induced cardiomyocyte pyroptosis through the NLRP3/Caspase-1 pathway. Activation of NLRP3 pathway reversed the protection of IP3R1 inhibition/ERP44 overexpression/Nifedipine D6 on H/R-induced cells. Overall, ERP44 binding to IP3R1 inhibits Ca2+ overload, thus alleviating pyroptosis and MI/R injury.
Collapse
Affiliation(s)
- Guixi Mo
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Xin Liu
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Yiyue Zhong
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Jian Mo
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Zhiyi Li
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Daheng Li
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Liangqing Zhang
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| | - Yijun Liu
- grid.410560.60000 0004 1760 3078Department of Anesthesiology, Affiliated Hospital of Guangdong Medical university, Zhanjiang, Guangdong P.R. China
| |
Collapse
|
12
|
The mystery of mitochondria-ER contact sites in physiology and pathology: A cancer perspective. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165834. [PMID: 32437958 DOI: 10.1016/j.bbadis.2020.165834] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria-associated membranes (MAM), physical platforms that enable communication between mitochondria and the endoplasmic reticulum (ER), are enriched with many proteins and enzymes involved in several crucial cellular processes, such as calcium (Ca2+) homeostasis, lipid synthesis and trafficking, autophagy and reactive oxygen species (ROS) production. Accumulating studies indicate that tumor suppressors and oncogenes are present at these intimate contacts between mitochondria and the ER, where they influence Ca2+ flux between mitochondria and the ER or affect lipid homeostasis at MAM, consequently impacting cell metabolism and cell fate. Understanding these fundamental roles of mitochondria-ER contact sites as important domains for tumor suppressors and oncogenes can support the search for new and more precise anticancer therapies. In the present review, we summarize the current understanding of basic MAM biology, composition and function and discuss the possible role of MAM-resident oncogenes and tumor suppressors.
Collapse
|
13
|
Lee SH, Hadipour-Lakmehsari S, Murthy HR, Gibb N, Miyake T, Teng ACT, Cosme J, Yu JC, Moon M, Lim S, Wong V, Liu P, Billia F, Fernandez-Gonzalez R, Stagljar I, Sharma P, Kislinger T, Scott IC, Gramolini AO. REEP5 depletion causes sarco-endoplasmic reticulum vacuolization and cardiac functional defects. Nat Commun 2020; 11:965. [PMID: 32075961 PMCID: PMC7031342 DOI: 10.1038/s41467-019-14143-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
The sarco-endoplasmic reticulum (SR/ER) plays an important role in the development and progression of many heart diseases. However, many aspects of its structural organization remain largely unknown, particularly in cells with a highly differentiated SR/ER network. Here, we report a cardiac enriched, SR/ER membrane protein, REEP5 that is centrally involved in regulating SR/ER organization and cellular stress responses in cardiac myocytes. In vitro REEP5 depletion in mouse cardiac myocytes results in SR/ER membrane destabilization and luminal vacuolization along with decreased myocyte contractility and disrupted Ca2+ cycling. Further, in vivo CRISPR/Cas9-mediated REEP5 loss-of-function zebrafish mutants show sensitized cardiac dysfunction upon short-term verapamil treatment. Additionally, in vivo adeno-associated viral (AAV9)-induced REEP5 depletion in the mouse demonstrates cardiac dysfunction. These results demonstrate the critical role of REEP5 in SR/ER organization and function as well as normal heart function and development.
Collapse
Affiliation(s)
- Shin-Haw Lee
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Sina Hadipour-Lakmehsari
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Harsha R Murthy
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Natalie Gibb
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Tetsuaki Miyake
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biology, Faculty of Science, York University, Toronto, ON, M3J1P3, Canada
| | - Allen C T Teng
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Jake Cosme
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Jessica C Yu
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
| | - Mark Moon
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - SangHyun Lim
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto Canada, Toronto, Canada
| | - Victoria Wong
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Peter Liu
- Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Filio Billia
- Toronto General Research Institute, University Health Network, Toronto, ON, M5G2C4, Canada
| | - Rodrigo Fernandez-Gonzalez
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, M5S3G9, Canada
| | - Igor Stagljar
- Donnelly Centre, University of Toronto, Toronto, ON, M5S1M8, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto Canada, Toronto, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, Canada
- Mediterranean Institute for Life Sciences, Split, Croatia
| | - Parveen Sharma
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
- Institute of Translational Medicine, University of Liverpool, Liverpool, L693BX, UK
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, Toronto, ON, M5G1L7, Canada
| | - Ian C Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, M5G1M1, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, M5S1M8, Canada.
| |
Collapse
|
14
|
The role of ERp44 in glucose and lipid metabolism. Arch Biochem Biophys 2019; 671:175-184. [PMID: 31283909 DOI: 10.1016/j.abb.2019.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
Endoplasmic Reticulum Protein 44 (ERp44) is a member of the PDI family, named for a molecular weight of 44 kD. White adipose tissue has metabolic and endocrine functions that are important to metabolism. The role of ERp44 in glucose and lipid metabolism is not known yet. The current study was undertaken to investigate the implication of ERp44 in glucose and lipid metabolism. In this study, we generated and characterized ERp44-/- mice. We used type 2 diabetes models and ERp44 knockout mice to show the implication of ERp44 in glucose and lipid metabolism. Knockout newborns had lower blood glucose compared to wild-type. Adult knockouts had abnormal intraperitoneal, glucose, insulin and pyruvic acid tolerance. Lipocytes were smaller and fewer in knockout mice compared to wild-type. Knockouts resisted to high-fat diet-induced obesity. ERp44 expression in white adipose tissue decreased significantly in type 2 diabetes models. Results suggest that ERp44 is closely associated with glucose and lipid metabolism.
Collapse
|
15
|
The Best for the Most Important: Maintaining a Pristine Proteome in Stem and Progenitor Cells. Stem Cells Int 2019; 2019:1608787. [PMID: 31191665 PMCID: PMC6525796 DOI: 10.1155/2019/1608787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells give rise to reproductively enabled offsprings by generating progressively lineage-restricted multipotent stem cells that would differentiate into lineage-committed stem and progenitor cells. These lineage-committed stem and progenitor cells give rise to all adult tissues and organs. Adult stem and progenitor cells are generated as part of the developmental program and play critical roles in tissue and organ maintenance and/or regeneration. The ability of pluripotent stem cells to self-renew, maintain pluripotency, and differentiate into a multicellular organism is highly dependent on sensing and integrating extracellular and extraorganismal cues. Proteins perform and integrate almost all cellular functions including signal transduction, regulation of gene expression, metabolism, and cell division and death. Therefore, maintenance of an appropriate mix of correctly folded proteins, a pristine proteome, is essential for proper stem cell function. The stem cells' proteome must be pristine because unfolded, misfolded, or otherwise damaged proteins would interfere with unlimited self-renewal, maintenance of pluripotency, differentiation into downstream lineages, and consequently with the development of properly functioning tissue and organs. Understanding how various stem cells generate and maintain a pristine proteome is therefore essential for exploiting their potential in regenerative medicine and possibly for the discovery of novel approaches for maintaining, propagating, and differentiating pluripotent, multipotent, and adult stem cells as well as induced pluripotent stem cells. In this review, we will summarize cellular networks used by various stem cells for generation and maintenance of a pristine proteome. We will also explore the coordination of these networks with one another and their integration with the gene regulatory and signaling networks.
Collapse
|
16
|
Wen XY, Tarailo-Graovac M, Brand-Arzamendi K, Willems A, Rakic B, Huijben K, Da Silva A, Pan X, El-Rass S, Ng R, Selby K, Philip AM, Yun J, Ye XC, Ross CJ, Lehman AM, Zijlstra F, Abu Bakar N, Drögemöller B, Moreland J, Wasserman WW, Vallance H, van Scherpenzeel M, Karbassi F, Hoskings M, Engelke U, de Brouwer A, Wevers RA, Pshezhetsky AV, van Karnebeek CD, Lefeber DJ. Sialic acid catabolism by N-acetylneuraminate pyruvate lyase is essential for muscle function. JCI Insight 2018; 3:122373. [PMID: 30568043 DOI: 10.1172/jci.insight.122373] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/14/2018] [Indexed: 11/17/2022] Open
Abstract
Sialic acids are important components of glycoproteins and glycolipids essential for cellular communication, infection, and metastasis. The importance of sialic acid biosynthesis in human physiology is well illustrated by the severe metabolic disorders in this pathway. However, the biological role of sialic acid catabolism in humans remains unclear. Here, we present evidence that sialic acid catabolism is important for heart and skeletal muscle function and development in humans and zebrafish. In two siblings, presenting with sialuria, exercise intolerance/muscle wasting, and cardiac symptoms in the brother, compound heterozygous mutations [chr1:182775324C>T (c.187C>T; p.Arg63Cys) and chr1:182772897A>G (c.133A>G; p.Asn45Asp)] were found in the N-acetylneuraminate pyruvate lyase gene (NPL). In vitro, NPL activity and sialic acid catabolism were affected, with a cell-type-specific reduction of N-acetyl mannosamine (ManNAc). A knockdown of NPL in zebrafish resulted in severe skeletal myopathy and cardiac edema, mimicking the human phenotype. The phenotype was rescued by expression of wild-type human NPL but not by the p.Arg63Cys or p.Asn45Asp mutants. Importantly, the myopathy phenotype in zebrafish embryos was rescued by treatment with the catabolic products of NPL: N-acetyl glucosamine (GlcNAc) and ManNAc; the latter also rescuing the cardiac phenotype. In conclusion, we provide the first report to our knowledge of a human defect in sialic acid catabolism, which implicates an important role of the sialic acid catabolic pathway in mammalian muscle physiology, and suggests opportunities for monosaccharide replacement therapy in human patients.
Collapse
Affiliation(s)
- Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Maja Tarailo-Graovac
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Institute of Physiology and Biochemistry, Faculty of Biology, The University of Belgrade, Belgrade, Serbia.,Departments of Biochemistry, Molecular Biology, and Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Koroboshka Brand-Arzamendi
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Anke Willems
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bojana Rakic
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Karin Huijben
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Afitz Da Silva
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Xuefang Pan
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Robin Ng
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Katheryn Selby
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Anju Mary Philip
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Junghwa Yun
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - X Cynthia Ye
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Colin J Ross
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Anna M Lehman
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Fokje Zijlstra
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - N Abu Bakar
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Britt Drögemöller
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver Canada
| | - Jacqueline Moreland
- Departments of Biochemistry, Molecular Biology, and Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Wyeth W Wasserman
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Hilary Vallance
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Monique van Scherpenzeel
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands.,Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Farhad Karbassi
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Martin Hoskings
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver Canada
| | - Udo Engelke
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arjan de Brouwer
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alexey V Pshezhetsky
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Clara Dm van Karnebeek
- Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver Canada.,Departments of Pediatrics and Clinical Genetics, Emma Children's Hospital, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands.,Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
17
|
ERp44 depletion exacerbates ER stress and aggravates diabetic nephropathy in db/db mice. Biochem Biophys Res Commun 2018; 504:921-926. [PMID: 30224065 DOI: 10.1016/j.bbrc.2018.09.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/07/2018] [Indexed: 12/25/2022]
Abstract
Diabetic nephropathy (DN) is a major complication of diabetes, and the dysfunction of endoplasmic reticulum (ER) plays an important role in its pathogenesis. ERp44, an ER resident chaperone protein, has been implicated in the modulation of ER stress, however, its role and mechanism in DN are not determined. Here, we show that ERp44 expression is upregulated in the glomeruli of db/db mice, a rodent model of type 2 diabetes. When ERp44 is depleted by in vivo shRNA-mediated knockdown, the features associated with DN including albuminuria level and glomerular basement membrane (GBM) thickness are aggravated, therefore suggesting a detrimental role of ERp44 depletion in DN progression. We further show that ERp44 depletion exacerbates ER stress in DN in db/db mice, and that attenuating ER stress with the chemical chaperone TUDCA remarkably diminishes the aggravated DN features caused by ERp44 depletion. These results suggest that the exacerbated ER stress is a critical factor for the detrimental effect of ERp44 depletion on DN progression in db/db mice. Thus, our study links the role of ERp44 in DN with ER stress regulation and may offer a potential therapeutic strategy to interfere DN progression.
Collapse
|
18
|
Massé S, Magtibay K, Jackson N, Asta J, Kusha M, Zhang B, Balachandran R, Radisic M, Deno DC, Nanthakumar K. Resolving Myocardial Activation With Novel Omnipolar Electrograms. Circ Arrhythm Electrophysiol 2017; 9:e004107. [PMID: 27406608 PMCID: PMC4956680 DOI: 10.1161/circep.116.004107] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/01/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND With its inherent limitations, determining local activation times has been the basis of cardiac mapping for over a century. Here, we introduce omnipolar electrograms that originate from the natural direction of a travelling wave and from which instantaneous conduction velocity amplitude and direction can be computed at any single location without first determining activation times. We sought to validate omnipole-derived conduction velocities and explore potential application for localization of sources of arrhythmias. METHODS AND RESULTS Electrograms from omnipolar mapping were derived and validated using 4 separate models and 2 independent signal acquisition methodologies. We used both electric signals and optical signals collected from monolayer cell preparations, 3-dimensional constructs built with cardiomyocytes derived from human embryonic stem cells, simultaneous optical and electric mapping of rabbit hearts, and in vivo pig electrophysiology studies. Conduction velocities calculated from omnipolar electrograms were compared with wavefront propagation from optical and electric-mapping studies with a traditional local activation time-based method. Bland-Altman analysis revealed that omnipolar measurements on optical data were in agreement with local activation time methods for wavefront direction and velocity within 25 cm/s and 30°, respectively. Similar agreement was also found on electric data. Furthermore, mathematical operations, such as curl and divergence, were applied to omnipole-derived velocity vector fields to locate rotational and focal sources, respectively. CONCLUSIONS Electrode orientation-independent cardiac wavefront trajectory and speed at a single location for each cardiac activation can be determined accurately with omnipolar electrograms. Omnipole-derived vector fields, when combined with mathematical transforms may aid in real-time detection of cardiac activation sources.
Collapse
Affiliation(s)
- Stéphane Massé
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Karl Magtibay
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Nicholas Jackson
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - John Asta
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Marjan Kusha
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Boyang Zhang
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Ram Balachandran
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Milica Radisic
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - D Curtis Deno
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| | - Kumaraswamy Nanthakumar
- From the The Hull Family Cardiac Fibrillation Management Laboratory and University Health Network, Toronto General Hospital, Toronto, ON, Canada (S.M., K.M., N.J., J.A., M.K., K.N.); St. Jude Medical, St. Paul, MN (R.B., D.C.D.); and Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada (B.Z., M.R.)
| |
Collapse
|
19
|
Gutiérrez T, Simmen T. Endoplasmic reticulum chaperones tweak the mitochondrial calcium rheostat to control metabolism and cell death. Cell Calcium 2017; 70:64-75. [PMID: 28619231 DOI: 10.1016/j.ceca.2017.05.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 12/16/2022]
Abstract
The folding of secretory proteins is a well-understood mechanism, based on decades of research on endoplasmic reticulum (ER) chaperones. These chaperones interact with newly imported polypeptides close to the ER translocon. Classic examples for these proteins include the immunoglobulin binding protein (BiP/GRP78), and the lectins calnexin and calreticulin. Although not considered chaperones per se, the ER oxidoreductases of the protein disulfide isomerase (PDI) family complete the folding job by catalyzing the formation of disulfide bonds through cysteine oxidation. Research from the past decade has demonstrated that ER chaperones are multifunctional proteins. The regulation of ER-mitochondria Ca2+ crosstalk is one of their additional functions, as shown for calnexin, BiP/GRP78 or the oxidoreductases Ero1α and TMX1. This function depends on interactions of this group of proteins with the ER Ca2+ handling machinery. This novel function makes perfect sense for two reasons: i. It allows ER chaperones to control mitochondrial apoptosis instantly without a lengthy bypass involving the upregulation of pro-apoptotic transcription factors via the unfolded protein response (UPR); and ii. It allows the ER protein folding machinery to fine-tune ATP import via controlling the speed of mitochondrial oxidative phosphorylation. Therefore, the role of ER chaperones in regulating ER-mitochondria Ca2+ flux identifies the progression of secretory protein folding as a central regulator of cell survival and death, at least in cell types that secrete large amount of proteins. In other cell types, ER protein folding might serve as a sentinel mechanism that monitors cellular well-being to control cell metabolism and apoptosis. The selenoprotein SEPN1 is a classic example for such a role. Through the control of ER-mitochondria Ca2+-flux, ER chaperones and folding assistants guide cellular apoptosis and mitochondrial metabolism.
Collapse
Affiliation(s)
- Tomas Gutiérrez
- Faculty of Medicine and Dentistry, Department of Cell Biology, University of Alberta, Edmonton, T6G2H7, Canada
| | - Thomas Simmen
- Faculty of Medicine and Dentistry, Department of Cell Biology, University of Alberta, Edmonton, T6G2H7, Canada,.
| |
Collapse
|
20
|
Crystal Structure of the ERp44-Peroxiredoxin 4 Complex Reveals the Molecular Mechanisms of Thiol-Mediated Protein Retention. Structure 2016; 24:1755-1765. [PMID: 27642162 DOI: 10.1016/j.str.2016.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/22/2016] [Accepted: 08/05/2016] [Indexed: 12/23/2022]
Abstract
ERp44 controls the localization and transport of diverse proteins in the early secretory pathway. The mechanisms that allow client recognition and the source of the oxidative power for forming intermolecular disulfides are as yet unknown. Here we present the structure of ERp44 bound to a client, peroxiredoxin 4. Our data reveal that ERp44 binds the oxidized form of peroxiredoxin 4 via thiol-disulfide interchange reactions. The structure explains the redox-dependent recognition and characterizes the essential non-covalent interactions at the interface. The ERp44-Prx4 covalent complexes can be reduced by glutathione and protein disulfide isomerase family members in the ER, allowing the two components to recycle. This work provides insights into the mechanisms of thiol-mediated protein retention and indicates the key roles of ERp44 in this biochemical cycle to optimize oxidative folding and redox homeostasis.
Collapse
|
21
|
Yang Y, Cheung HH, Tu J, Miu KK, Chan WY. New insights into the unfolded protein response in stem cells. Oncotarget 2016; 7:54010-54027. [PMID: 27304053 PMCID: PMC5288239 DOI: 10.18632/oncotarget.9833] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/29/2016] [Indexed: 12/15/2022] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive mechanism to increase cell survival under endoplasmic reticulum (ER) stress conditions. The UPR is critical for maintaining cell homeostasis under physiological and pathological conditions. The vital functions of the UPR in development, metabolism and immunity have been demonstrated in several cell types. UPR dysfunction activates a variety of pathologies, including cancer, inflammation, neurodegenerative disease, metabolic disease and immune disease. Stem cells with the special ability to self-renew and differentiate into various somatic cells have been demonstrated to be present in multiple tissues. These cells are involved in development, tissue renewal and certain disease processes. Although the role and regulation of the UPR in somatic cells has been widely reported, the function of the UPR in stem cells is not fully known, and the roles and functions of the UPR are dependent on the stem cell type. Therefore, in this article, the potential significances of the UPR in stem cells, including embryonic stem cells, tissue stem cells, cancer stem cells and induced pluripotent cells, are comprehensively reviewed. This review aims to provide novel insights regarding the mechanisms associated with stem cell differentiation and cancer pathology.
Collapse
Affiliation(s)
- Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan, Ningxia, P.R. China
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Hoi Hung Cheung
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - JiaJie Tu
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Kai Kei Miu
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| | - Wai Yee Chan
- The Chinese University of Hong Kong–Shandong University Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, HKSAR, China
| |
Collapse
|
22
|
ERp44 Exerts Redox-Dependent Control of Blood Pressure at the ER. Mol Cell 2015; 58:1015-27. [DOI: 10.1016/j.molcel.2015.04.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 01/16/2015] [Accepted: 03/31/2015] [Indexed: 01/09/2023]
|
23
|
Anelli T, Sannino S, Sitia R. Proteostasis and "redoxtasis" in the secretory pathway: Tales of tails from ERp44 and immunoglobulins. Free Radic Biol Med 2015; 83:323-30. [PMID: 25744412 DOI: 10.1016/j.freeradbiomed.2015.02.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/20/2015] [Accepted: 02/22/2015] [Indexed: 01/09/2023]
Abstract
In multicellular organisms, some cells are given the task of secreting huge quantities of proteins. To comply with their duty, they generally equip themselves with a highly developed endoplasmic reticulum (ER) and downstream organelles in the secretory pathway. These professional secretors face paramount proteostatic challenges in that they need to couple efficiency and fidelity in their secretory processes. On one hand, stringent quality control (QC) mechanisms operate from the ER onward to check the integrity of the secretome. On the other, the pressure to secrete can be overwhelming, as for instance on antibody-producing cells during infection. Maintaining homeostasis is particularly hard when the products to be released contain disulfide bonds, because oxidative folding entails production of reactive oxygen species. How are redox homeostasis ("redoxtasis") and proteostasis maintained despite the massive fluxes of cargo proteins traversing the pathway? Here we describe recent findings on how ERp44, a multifunctional chaperone of the secretory pathway, can modulate these processes integrating protein QC, redoxtasis, and calcium signaling.
Collapse
Affiliation(s)
- Tiziana Anelli
- Divisions of Genetics and Cell Biology, IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Sara Sannino
- Divisions of Genetics and Cell Biology, IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Roberto Sitia
- Divisions of Genetics and Cell Biology, IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|