1
|
Xue R, Li M, Zhang G, Zhang W, Han L, Bo T, Zhong H, Yao D, Deng Y, Chen S, Zhang S. GSDME-mediated pyroptosis contributes to chemotherapy-induced platelet hyperactivity and thrombotic potential. Blood 2024; 144:2652-2665. [PMID: 39378585 DOI: 10.1182/blood.2023023179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 10/10/2024] Open
Abstract
ABSTRACT Thrombotic complications due to platelet hyperreactivity are a major cause of death in patients undergoing chemotherapy. However, the underlying mechanisms are not fully understood. Herein, using human platelets and platelets from mice lacking gasdermin E (GSDME), we show that GSDME is functionally expressed in anucleate platelets, and that GSDME-mediated pyroptosis, a newly identified form of cell death in mammalian nucleated cells, contributes to platelet hyperactivity in cisplatin-based chemotherapy. Cisplatin or etoposide activates caspase-3 to cleave GSDME, thereby releasing the N-terminal fragment of GSDME (GSDME-N) toward the platelet plasma membrane, subsequently forming membrane pores and facilitating platelet granule release. This eventually promotes platelet hyperactivity and thrombotic potential. We identified flotillin-2, a scaffold protein, as a GSDME-N interactor that recruits GSDME-N to the platelet membrane. Loss of GSDME protects mice from cisplatin-induced platelet hyperactivity. Our results provide evidence that targeting GSDME-mediated pyroptosis could reduce thrombotic potential in chemotherapy.
Collapse
Affiliation(s)
- Ruyi Xue
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Li
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ge Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liping Han
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Tao Bo
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Haoxuan Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Dingjin Yao
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiran Deng
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - She Chen
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Ayuyan AG, Cherny VV, Chaves G, Musset B, Cohen FS, DeCoursey TE. Interaction with stomatin directs human proton channels into cholesterol-dependent membrane domains. Biophys J 2024; 123:4180-4190. [PMID: 38444158 DOI: 10.1016/j.bpj.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/24/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024] Open
Abstract
Many membrane proteins are modulated by cholesterol. Here we report profound effects of cholesterol depletion and restoration on the human voltage-gated proton channel, hHV1, in excised patches but negligible effects in the whole-cell configuration. Despite the presence of a putative cholesterol-binding site, a CARC motif in hHV1, mutation of this motif did not affect cholesterol effects. The murine HV1 lacks a CARC sequence but displays similar cholesterol effects. These results argue against a direct effect of cholesterol on the HV1 protein. However, the data are fully explainable if HV1 preferentially associates with cholesterol-dependent lipid domains, or "rafts." The rafts would be expected to concentrate in the membrane/glass interface and to be depleted from the electrically accessible patch membrane. This idea is supported by evidence that HV1 channels can diffuse between seal and patch membranes when suction is applied. Simultaneous truncation of the large intracellular N and C termini of hHV1 greatly attenuated the cholesterol effect, but C truncation alone did not; this suggests that the N terminus is the region of attachment to lipid domains. Searching for abundant raft-associated proteins led to stomatin. Co-immunoprecipitation experiment results were consistent with hHV1 binding to stomatin. The stomatin-mediated association of HV1 with cholesterol-dependent lipid domains provides a mechanism for cells to direct HV1 to subcellular locations where it is needed, such as the phagosome in leukocytes.
Collapse
Affiliation(s)
- Artem G Ayuyan
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois.
| | - Vladimir V Cherny
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois
| | - Gustavo Chaves
- Institut für Physiologie, Pathophysiologie und Biophysik, CPPB, Paracelsus Medical University, Nürnberg, Germany
| | - Boris Musset
- Institut für Physiologie, Pathophysiologie und Biophysik, CPPB, Paracelsus Medical University, Nürnberg, Germany
| | - Fredric S Cohen
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois
| | - Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois.
| |
Collapse
|
3
|
Manchanda Y, ElEid L, Oqua AI, Ramchunder Z, Choi J, Shchepinova MM, Rutter GA, Inoue A, Tate EW, Jones B, Tomas A. Engineered mini-G proteins block the internalization of cognate GPCRs and disrupt downstream intracellular signaling. Sci Signal 2024; 17:eabq7038. [PMID: 38954638 DOI: 10.1126/scisignal.abq7038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
Mini-G proteins are engineered, thermostable variants of Gα subunits designed to stabilize G protein-coupled receptors (GPCRs) in their active conformations. Because of their small size and ease of use, they are popular tools for assessing GPCR behaviors in cells, both as reporters of receptor coupling to Gα subtypes and for cellular assays to quantify compartmentalized signaling at various subcellular locations. Here, we report that overexpression of mini-G proteins with their cognate GPCRs disrupted GPCR endocytic trafficking and associated intracellular signaling. In cells expressing the Gαs-coupled GPCR glucagon-like peptide 1 receptor (GLP-1R), coexpression of mini-Gs, a mini-G protein derived from Gαs, blocked β-arrestin 2 recruitment and receptor internalization and disrupted endosomal GLP-1R signaling. These effects did not involve changes in receptor phosphorylation or lipid nanodomain segregation. Moreover, we found that mini-G proteins derived from Gαi and Gαq also inhibited the internalization of GPCRs that couple to them. Finally, we developed an alternative intracellular signaling assay for GLP-1R using a nanobody specific for active Gαs:GPCR complexes (Nb37) that did not affect GLP-1R internalization. Our results have important implications for designing methods to assess intracellular GPCR signaling.
Collapse
Affiliation(s)
- Yusman Manchanda
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Liliane ElEid
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Affiong I Oqua
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Zenouska Ramchunder
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Jiyoon Choi
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Maria M Shchepinova
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
- CR-CHUM, Université de Montréal, Montréal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, London, UK
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, UK
| |
Collapse
|
4
|
Hagiyama M, Yoneshige A, Wada A, Kimura R, Ito S, Inoue T, Takeuchi F, Ito A. Efficient intracellular drug delivery by co-administration of two antibodies against cell adhesion molecule 1. J Control Release 2024; 371:603-618. [PMID: 38782061 DOI: 10.1016/j.jconrel.2024.05.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Cell adhesion molecule 1 (CADM1), a single-pass transmembrane protein, is involved in oncogenesis. We previously demonstrated the therapeutic efficacy of anti-CADM1 ectodomain monoclonal antibodies against mesothelioma; however, the underlying mechanism is unclear. In the present study, we explored the molecular behavior of anti-CADM1 antibodies in CADM1-expressing tumor cells. Sequencing analyses revealed that the anti-CADM1 chicken monoclonal antibodies 3E1 and 9D2 are IgY and IgM isotype antibodies, respectively. Co-administration of 3E1 and 9D2 altered the subcellular distribution of CADM1 from the detergent-soluble fraction to the detergent-resistant fraction in tumor cells. Using recombinant chicken-mouse chimeric antibodies that had been isotype-switched from IgG to IgM, we demonstrated that the combination of the variable region of 3E1 and the constant region of IgM was required for CADM1 relocation. Cytochemical studies showed that 3E1 colocalized with late endosomes/lysosomes after co-administration with 9D2, suggesting that the CADM1-antibody complex is internalized from the cell surface to intracellular compartments by lipid-raft mediated endocytosis. Finally, 3E1 was conjugated with the antimitotic agent monomethyl auristatin E (MMAE) via a cathepsin-cleavable linker. Co-administration of 3E1-monomethyl auristatin E and 9D2 suppressed the growth of multiple types of tumor cells, and this anti-tumor activity was confirmed in a syngeneic mouse model of melanoma. 3E1 and 9D2 are promising drug delivery vehicles for CADM1-expressing tumor cells.
Collapse
Affiliation(s)
- Man Hagiyama
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan.
| | - Akihiro Wada
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Ryuichiro Kimura
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takao Inoue
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Fuka Takeuchi
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Akihiko Ito
- Department of Pathology, Faculty of Medicine, Kindai University, Osaka, Japan.
| |
Collapse
|
5
|
Dey S, Mohapatra S, Khokhar M, Hassan S, Pandey RK. Extracellular Vesicles in Malaria: Shedding Light on Pathogenic Depths. ACS Infect Dis 2024; 10:827-844. [PMID: 38320272 PMCID: PMC10928723 DOI: 10.1021/acsinfecdis.3c00649] [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: 11/26/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/08/2024]
Abstract
Malaria, a life-threatening infectious disease caused by Plasmodium falciparum, remains a significant global health challenge, particularly in tropical and subtropical regions. The epidemiological data for 2021 revealed a staggering toll, with 247 million reported cases and 619,000 fatalities attributed to the disease. This formidable global health challenge continues to perplex researchers seeking a comprehensive understanding of its pathogenesis. Recent investigations have unveiled the pivotal role of extracellular vesicles (EVs) in this intricate landscape. These tiny, membrane-bound vesicles, secreted by diverse cells, emerge as pivotal communicators in malaria's pathogenic orchestra. This Review delves into the multifaceted roles of EVs in malaria pathogenesis, elucidating their impact on disease progression and immune modulation. Insights into EV involvement offer potential therapeutic and diagnostic strategies. Integrating this information identifies targets to mitigate malaria's global impact. Moreover, this Review explores the potential of EVs as diagnostic biomarkers and therapeutic targets in malaria. By deciphering the intricate dialogue facilitated by these vesicles, new avenues for intervention and novel strategies for disease management may emerge.
Collapse
Affiliation(s)
- Sangita Dey
- CSO
Department, Cellworks Research India Pvt
Ltd, Bengaluru 560066, Karnataka, India
| | - Salini Mohapatra
- Department
of Biotechnology, Chandigarh University, Punjab 140413, India
| | - Manoj Khokhar
- Department
of Biochemistry, All India Institute of
Medical Sciences Jodhpur, Rajasthan 342005, India
| | - Sana Hassan
- Department
of Life Sciences, Manipal Academy of Higher
Education, Dubai 345050, United Arab Emirates
| | - Rajan Kumar Pandey
- Department
of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| |
Collapse
|
6
|
Salzer U, Mairhofer M, De Franceschi L. Rainer Prohaska (1943-2022). Am J Hematol 2024; 99:144-145. [PMID: 37867398 DOI: 10.1002/ajh.27139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Affiliation(s)
- Ulrich Salzer
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Mario Mairhofer
- Medical Department of Hematology and Oncology, Johannes Kepler University, Linz, Austria
| | | |
Collapse
|
7
|
Leinung N, Mentrup T, Patel M, Gallagher T, Schröder B. Dynamic association of the intramembrane proteases SPPL2a/b and their substrates with tetraspanin-enriched microdomains. iScience 2023; 26:107819. [PMID: 37736044 PMCID: PMC10509304 DOI: 10.1016/j.isci.2023.107819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/21/2023] [Accepted: 08/31/2023] [Indexed: 09/23/2023] Open
Abstract
Signal peptide peptidase-like 2a and b (SPPL2a/b) are aspartyl intramembrane proteases and cleave tail-anchored proteins as well as N-terminal fragments (NTFs) derived from type II-oriented transmembrane proteins. How these proteases recruit substrates and cleavage is regulated, is still incompletely understood. We found that SPPL2a/b localize to detergent-resistant membrane (DRM) domains with the characteristics of tetraspanin-enriched microdomains (TEMs). Based on this, association with several tetraspanins was evaluated. We demonstrate that not only SPPL2a/b but also their substrates tumor necrosis factor (TNF) and CD74 associate with tetraspanins like CD9, CD81, and CD82 and/or TEMs and analyze the stability of these complexes in different detergents. CD9 and CD81 deficiency has protease- and substrate-selective effects on SPPL2a/b function. Our findings suggest that reciprocal interactions with tetraspanins may assist protease-substrate encounters of SPPL2a/b within the membrane. Beyond SPP/SPPL proteases, this supports previous concepts that tetraspanins facilitate membrane-embedded proteolytic processes.
Collapse
Affiliation(s)
- Nadja Leinung
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Torben Mentrup
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Mehul Patel
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL, USA
| | - Bernd Schröder
- Institute of Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
8
|
Wang Z, Zhou X, Deng X, Ye D, Liu D, Zhou B, Zheng W, Wang X, Wang Y, Borkhuu O, Fang L. miR-186-ANXA9 signaling inhibits tumorigenesis in breast cancer. Front Oncol 2023; 13:1166666. [PMID: 37841425 PMCID: PMC10570552 DOI: 10.3389/fonc.2023.1166666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Breast cancer (BC) ranks as the highest incidence among cancer types in women all over the world. MicroRNAs (miRNAs) are a class of short endogenous non-coding RNA in cells mostly functioning to silence the target mRNAs. In the current study, a miRNA screening analysis identified miR-186-5p to be downregulated in human breast cancer tumors. Functional studies in vitro demonstrated that overexpression of miR-186-5p inhibited cellular proliferation and induced cell apoptosis in multiple breast cancer cell lines including MDA-MB-231, MCF-7, and BT549 cells. Transplantation of the miR-186-5p-overexpressing MDA-MB-231 cells into nude mice significantly inhibited mammary tumor growth in vivo. Sequence blast analysis predicted annexin A9 (ANXA9) as a target gene of miR-186-5p, which was validated by luciferase reporter assay, QRT-PCR analysis, and western blot. Additional gene expression analysis of clinical tumor samples indicated a negative correlation between miR-186-5p and ANXA9 in human breast cancer. Knockdown of ANXA9 mimicked the phenotype of miR-186-5p overexpression. Reintroduction of ANXA9 back rescued the miR-186-5p-induced cell apoptosis. In addition, miR-186-5p decreased the expression of Bcl-2 and increased the expression of p53, suggesting a mechanism regulating miR-186-5p-induced cellular apoptosis. In summary, our study is the first to demonstrate miR-186-5p-ANXA9 signaling in suppressing human breast cancer. It provided a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Zhongrui Wang
- Department of Thyroid and Breast Surgery, Shanghai Tenth People’s Hospital, Shanghai Tenth People’s Hospital of Nanjing Medical University, Shanghai, China
- Department of Breast and Thyroid Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiqian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaochong Deng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danrong Ye
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Diya Liu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baian Zhou
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenfang Zheng
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuehui Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuying Wang
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Oyungerel Borkhuu
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Fang
- Department of Thyroid and Breast Surgery, Shanghai Tenth People’s Hospital, Shanghai Tenth People’s Hospital of Nanjing Medical University, Shanghai, China
- Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Marques-da-Silva D, Lagoa R. Rafting on the Evidence for Lipid Raft-like Domains as Hubs Triggering Environmental Toxicants' Cellular Effects. Molecules 2023; 28:6598. [PMID: 37764374 PMCID: PMC10536579 DOI: 10.3390/molecules28186598] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The plasma membrane lipid rafts are cholesterol- and sphingolipid-enriched domains that allow regularly distributed, sub-micro-sized structures englobing proteins to compartmentalize cellular processes. These membrane domains can be highly heterogeneous and dynamic, functioning as signal transduction platforms that amplify the local concentrations and signaling of individual components. Moreover, they participate in cell signaling routes that are known to be important targets of environmental toxicants affecting cell redox status and calcium homeostasis, immune regulation, and hormonal functions. In this work, the evidence that plasma membrane raft-like domains operate as hubs for toxicants' cellular actions is discussed, and suggestions for future research are provided. Several studies address the insertion of pesticides and other organic pollutants into membranes, their accumulation in lipid rafts, or lipid rafts' disruption by polychlorinated biphenyls (PCBs), benzo[a]pyrene (B[a]P), and even metals/metalloids. In hepatocytes, macrophages, or neurons, B[a]P, airborne particulate matter, and other toxicants caused rafts' protein and lipid remodeling, oxidative changes, or amyloidogenesis. Different studies investigated the role of the invaginated lipid rafts present in endothelial cells in mediating the vascular inflammatory effects of PCBs. Furthermore, in vitro and in vivo data strongly implicate raft-localized NADPH oxidases, the aryl hydrocarbon receptor, caveolin-1, and protein kinases in the toxic mechanisms of occupational and environmental chemicals.
Collapse
Affiliation(s)
- Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
10
|
Fan Q, Li M, Zhao W, Zhang K, Li M, Li W. Hyper α2,6-Sialylation Promotes CD4 + T-Cell Activation and Induces the Occurrence of Ulcerative Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302607. [PMID: 37424034 PMCID: PMC10502867 DOI: 10.1002/advs.202302607] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/23/2023] [Indexed: 07/11/2023]
Abstract
α2,6-sialylation, catalyzed by α2,6-sialyltransferase (ST6GAL1), plays a pivotal role in immune responses. However, the role of ST6GAL1 in the pathogenesis of ulcerative colitis (UC) remains unknown. ST6GAL1 mRNA is highly expressed in UC tissues compared with the corresponding adjacent normal tissues, and α2,6-sialylation is significantly increased in the colon tissues of patients with UC. The expression of ST6GAL1 and proinflammatory cytokines, such as interleukin (IL)-2, IL-6, IL-17, and interferon-gamma, is also increased. The number of CD4+ T cells increases in UC patients. St6gal1 gene knockout (St6gal1-/- ) rats are established by clustered regularly interspaced short palindromic repeats (CRISPR)-associated gene knockout system. St6gal1 deficiency reduces the levels of pro-inflammatory cytokines and alleviates colitis symptoms in UC model rats. Ablation of α2,6-sialylation inhibits the transport of the TCR to lipid rafts and suppresses CD4+ T-cell activation. The attenuation of TCR signaling downregulates the expression of NF-κB in ST6GAL1-/- CD4+ T-cells. Moreover, NF-κB could bind to the ST6GAL1 promoter to increase its transcription. Ablation of ST6GAL1 downregulates the expression of NF-κB and reduces the production of proinflammatory cytokines to relieve UC pathogenesis, which is a potential novel target for the clinical treatment of UC.
Collapse
Affiliation(s)
- Qingjie Fan
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular ImmunopathologyShantou University Medical CollegeShantouGuangdong515041China
- College of Basic Medical ScienceDalian Medical University9‐Western Section, Lvshun South RoadDalianLiaoning116044China
| | - Mechou Li
- College of Basic Medical ScienceDalian Medical University9‐Western Section, Lvshun South RoadDalianLiaoning116044China
| | - Weiwei Zhao
- College of Basic Medical ScienceDalian Medical University9‐Western Section, Lvshun South RoadDalianLiaoning116044China
| | - Kaixin Zhang
- College of Basic Medical ScienceDalian Medical University9‐Western Section, Lvshun South RoadDalianLiaoning116044China
| | - Ming Li
- College of Basic Medical ScienceDalian Medical University9‐Western Section, Lvshun South RoadDalianLiaoning116044China
| | - Wenzhe Li
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular ImmunopathologyShantou University Medical CollegeShantouGuangdong515041China
| |
Collapse
|
11
|
Barshtein G, Gural A, Arbell D, Barkan R, Livshits L, Pajic-Lijakovic I, Yedgar S. Red Blood Cell Deformability Is Expressed by a Set of Interrelated Membrane Proteins. Int J Mol Sci 2023; 24:12755. [PMID: 37628935 PMCID: PMC10454903 DOI: 10.3390/ijms241612755] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Red blood cell (RBC) deformability, expressing their ability to change their shape, allows them to minimize their resistance to flow and optimize oxygen delivery to the tissues. RBC with reduced deformability may lead to increased vascular resistance, capillary occlusion, and impaired perfusion and oxygen delivery. A reduction in deformability, as occurs during RBC physiological aging and under blood storage, is implicated in the pathophysiology of diverse conditions with circulatory disorders and anemias. The change in RBC deformability is associated with metabolic and structural alterations, mostly uncharacterized. To bridge this gap, we analyzed the membrane protein levels, using mass spectroscopy, of RBC with varying deformability determined by image analysis. In total, 752 membrane proteins were identified. However, deformability was positively correlated with the level of only fourteen proteins, with a highly significant inter-correlation between them. These proteins are involved in membrane rafting and/or the membrane-cytoskeleton linkage. These findings suggest that the reduction of deformability is a programmed (not arbitrary) process of remodeling and shedding of membrane fragments, possibly mirroring the formation of extracellular vesicles. The highly significant inter-correlation between the deformability-expressing proteins infers that the cell deformability can be assessed by determining the level of a few, possibly one, of them.
Collapse
Affiliation(s)
- Gregory Barshtein
- Department of Biochemistry, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Alexander Gural
- Blood Bank, Hadassah University Hospital, Jerusalem 9112001, Israel;
| | - Dan Arbell
- Pediatric Surgery, Hadassah University Hospital, Jerusalem 9112001, Israel;
| | - Refael Barkan
- Department of Digital Medical Technologies, Holon Institute of Technology, Holon 5810201, Israel;
| | - Leonid Livshits
- Red Blood Cell Research Group, Vetsuisse Faculty, Institute of Veterinary Physiology, University of Zurich, 8057 Zürich, Switzerland;
| | | | - Saul Yedgar
- Department of Biochemistry, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| |
Collapse
|
12
|
Ghodsi M, Cloos AS, Mozaheb N, Van Der Smissen P, Henriet P, Pierreux CE, Cellier N, Mingeot-Leclercq MP, Najdovski T, Tyteca D. Variability of extracellular vesicle release during storage of red blood cell concentrates is associated with differential membrane alterations, including loss of cholesterol-enriched domains. Front Physiol 2023; 14:1205493. [PMID: 37408586 PMCID: PMC10318158 DOI: 10.3389/fphys.2023.1205493] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Transfusion of red blood cell concentrates is the most common medical procedure to treat anaemia. However, their storage is associated with development of storage lesions, including the release of extracellular vesicles. These vesicles affect in vivo viability and functionality of transfused red blood cells and appear responsible for adverse post-transfusional complications. However, the biogenesis and release mechanisms are not fully understood. We here addressed this issue by comparing the kinetics and extents of extracellular vesicle release as well as red blood cell metabolic, oxidative and membrane alterations upon storage in 38 concentrates. We showed that extracellular vesicle abundance increased exponentially during storage. The 38 concentrates contained on average 7 × 1012 extracellular vesicles at 6 weeks (w) but displayed a ∼40-fold variability. These concentrates were subsequently classified into 3 cohorts based on their vesiculation rate. The variability in extracellular vesicle release was not associated with a differential red blood cell ATP content or with increased oxidative stress (in the form of reactive oxygen species, methaemoglobin and band3 integrity) but rather with red blood cell membrane modifications, i.e., cytoskeleton membrane occupancy, lateral heterogeneity in lipid domains and transversal asymmetry. Indeed, no changes were noticed in the low vesiculation group until 6w while the medium and the high vesiculation groups exhibited a decrease in spectrin membrane occupancy between 3 and 6w and an increase of sphingomyelin-enriched domain abundance from 5w and of phosphatidylserine surface exposure from 8w. Moreover, each vesiculation group showed a decrease of cholesterol-enriched domains associated with a cholesterol content increase in extracellular vesicles but at different storage time points. This observation suggested that cholesterol-enriched domains could represent a starting point for vesiculation. Altogether, our data reveal for the first time that the differential extent of extracellular vesicle release in red blood cell concentrates did not simply result from preparation method, storage conditions or technical issues but was linked to membrane alterations.
Collapse
Affiliation(s)
- Marine Ghodsi
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Anne-Sophie Cloos
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Negar Mozaheb
- Cellular and Molecular Pharmacology Unit, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Patrick Van Der Smissen
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Christophe E. Pierreux
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| | | | | | - Tomé Najdovski
- Service du Sang, Croix-Rouge de Belgique, Suarlée, Belgium
| | - Donatienne Tyteca
- Cell Biology Unit and Platform for Imaging Cells and Tissues, de Duve Institute, UCLouvain, Brussels, Belgium
| |
Collapse
|
13
|
Molecular Mechanisms and Pathophysiological Significance of Eryptosis. Int J Mol Sci 2023; 24:ijms24065079. [PMID: 36982153 PMCID: PMC10049269 DOI: 10.3390/ijms24065079] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Despite lacking the central apoptotic machinery, senescent or damaged RBCs can undergo an unusual apoptosis-like cell death, termed eryptosis. This premature death can be caused by, or a symptom of, a wide range of diseases. However, various adverse conditions, xenobiotics, and endogenous mediators have also been recognized as triggers and inhibitors of eryptosis. Eukaryotic RBCs are unique among their cell membrane distribution of phospholipids. The change in the RBC membrane composition of the outer leaflet occurs in a variety of diseases, including sickle cell disease, renal diseases, leukemia, Parkinson’s disease, and diabetes. Eryptotic erythrocytes exhibit various morphological alterations such as shrinkage, swelling, and increased granulation. Biochemical changes include cytosolic Ca2+ increase, oxidative stress, stimulation of caspases, metabolic exhaustion, and ceramide accumulation. Eryptosis is an effective mechanism for the elimination of dysfunctional erythrocytes due to senescence, infection, or injury to prevent hemolysis. Nevertheless, excessive eryptosis is associated with multiple pathologies, most notably anemia, abnormal microcirculation, and prothrombotic risk; all of which contribute to the pathogenesis of several diseases. In this review, we provide an overview of the molecular mechanisms, physiological and pathophysiological relevance of eryptosis, as well as the potential role of natural and synthetic compounds in modulating RBC survival and death.
Collapse
|
14
|
The Lipid Raft-Associated Protein Stomatin Is Required for Accumulation of Dectin-1 in the Phagosomal Membrane and for Full Activity of Macrophages against Aspergillus fumigatus. mSphere 2023; 8:e0052322. [PMID: 36719247 PMCID: PMC9942578 DOI: 10.1128/msphere.00523-22] [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] [Indexed: 02/01/2023] Open
Abstract
Alveolar macrophages belong to the first line of defense against inhaled conidia of the human-pathogenic fungus Aspergillus fumigatus. In lung alveoli, they contribute to phagocytosis and elimination of conidia. As a counterdefense, conidia have a gray-green pigment that enables them to survive in phagosomes of macrophages for some time. Previously, we showed that this conidial pigment interferes with the formation of flotillin-dependent lipid raft microdomains in the phagosomal membrane, thereby preventing the formation of functional phagolysosomes. Besides flotillins, stomatin is a major component of lipid rafts and can be targeted to the membrane. However, only limited information on stomatin is available, in particular on its role in defense against pathogens. To determine the function of this integral membrane protein, a stomatin-deficient macrophage line was generated by CRISPR/Cas9 gene editing. Immunofluorescence microscopy and flow cytometry revealed that stomatin contributes to the phagocytosis of conidia and is important for recruitment of the β-glucan receptor dectin-1 to both the cytoplasmic membrane and phagosomal membrane. In stomatin knockout cells, fusion of phagosomes and lysosomes, recruitment of the vATPase to phagosomes, and tumor necrosis factor alpha (TNF-α) levels were reduced when cells were infected with pigmentless conidia. Thus, our data suggest that stomatin is involved in maturation of phagosomes via fostering fusion of phagosomes with lysosomes. IMPORTANCE Stomatin is an integral membrane protein that contributes to the uptake of microbes, e.g., spores of the human-pathogenic fungus Aspergillus fumigatus. By generation of a stomatin-deficient macrophage line by advanced genetic engineering, we found that stomatin is involved in the recruitment of the β-glucan receptor dectin-1 to the phagosomal membrane of macrophages. Furthermore, stomatin is involved in maturation of phagosomes via fostering fusion of phagosomes with lysosomes. The data provide new insights on the important role of stomatin in the immune response against human-pathogenic fungi.
Collapse
|
15
|
Wang K, Li Z, Egini O, Wadgaonkar R, Jiang XC, Chen Y. Atomic force microscopy reveals involvement of the cell envelope in biomechanical properties of sickle erythrocytes. BMC Biol 2023; 21:31. [PMID: 36782158 PMCID: PMC9926656 DOI: 10.1186/s12915-023-01523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Intracellular hemoglobin polymerization has been supposed to be the major determinant for the elevated rigidity/stiffness of sickle erythrocytes from sickle cell anemia (SCA) patients. However, the contribution of the cell envelope remains unclear. RESULTS In this study, using atomic force microscopy (AFM), we compared the normal and sickled erythrocyte surfaces for stiffness and topography. AFM detected that sickle cells had a rougher surface and were stiffer than normal erythrocytes and that sickle cell ghosts had a rougher surface (for both outer and inner surfaces) and were thicker than normal ghosts, the latter implying a higher membrane-associated hemoglobin content/layer in the sickle cell envelope. Compared to healthy subjects, the SCA patients had lower plasma lipoprotein levels. AFM further revealed that a mild concentration of methyl-β-cyclodextrin (MβCD, a putative cholesterol-depleting reagent) could induce an increase in roughness of erythrocytes/ghosts and a decrease in thickness of ghosts for both normal and sickle cells, implying that MβCD can alter the cell envelope from outside (cholesterol in the plasma membrane) to inside (membrane-associated hemoglobin). More importantly, MβCD also caused a more significant decrease in stiffness of sickle cells than that of normal erythrocytes. CONCLUSIONS The data reveal that besides the cytosolic hemoglobin fibers, the cell envelope containing the membrane-associated hemoglobin also is involved in the biomechanical properties (e.g., stiffness and shape maintenance) of sickle erythrocytes.
Collapse
Affiliation(s)
- Kun Wang
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China
| | - Zhiqiang Li
- Department of Cell Biology, SUNY Health Sciences University, State University of New York, Brooklyn, NY, 11203, USA
| | - Ogechukwu Egini
- Division of Hematology and Oncology, Department of Medicine, SUNY Health Sciences University, State University of New York, Brooklyn, NY, 11203, USA
| | - Raj Wadgaonkar
- Department of Cell Biology, SUNY Health Sciences University, State University of New York, Brooklyn, NY, 11203, USA
- VA Medical Center, Brooklyn, NY11208, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Health Sciences University, State University of New York, Brooklyn, NY, 11203, USA.
- VA Medical Center, Brooklyn, NY11208, USA.
| | - Yong Chen
- Jiangxi Key Laboratory for Microscale Interdisciplinary Study, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi, 330031, People's Republic of China.
- Department of Cell Biology, SUNY Health Sciences University, State University of New York, Brooklyn, NY, 11203, USA.
| |
Collapse
|
16
|
Rougé S, Genetet S, Leal Denis MF, Dussiot M, Schwarzbaum PJ, Ostuni MA, Mouro-Chanteloup I. Mechanosensitive Pannexin 1 Activity Is Modulated by Stomatin in Human Red Blood Cells. Int J Mol Sci 2022; 23:ijms23169401. [PMID: 36012667 PMCID: PMC9409209 DOI: 10.3390/ijms23169401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Pannexin 1 (PANX1) was proposed to drive ATP release from red blood cells (RBCs) in response to stress conditions. Stomatin, a membrane protein regulating mechanosensitive channels, has been proposed to modulate PANX1 activity in non-erythroid cells. To determine whether stomatin modulates PANX1 activity in an erythroid context, we have (i) assessed the in situ stomatin-PANX1 interaction in RBCs, (ii) measured PANX1-stimulated activity in RBCs expressing stomatin or from OverHydrated Hereditary Stomatocytosis (OHSt) patients lacking stomatin, and in erythroid K562 cells invalidated for stomatin. Proximity Ligation Assay coupled with flow imaging shows 27.09% and 6.13% positive events in control and OHSt RBCs, respectively. The uptake of dyes 5(6)-Carboxyfluorescein (CF) and TO-PRO-3 was used to evaluate PANX1 activity. RBC permeability for CF is 34% and 11.8% in control and OHSt RBCs, respectively. PANX1 permeability for TO-PRO-3 is 35.72% and 18.42% in K562 stom+ and stom− clones, respectively. These results suggest an interaction between PANX1 and stomatin in human RBCs and show a significant defect in PANX1 activity in the absence of stomatin. Based on these results, we propose that stomatin plays a major role in opening the PANX1 pore by being involved in a caspase-independent lifting of autoinhibition.
Collapse
Affiliation(s)
- Sarah Rougé
- Université Paris Cité and Université des Antilles, INSERM U1134, BIGR, F-75014 Paris, France
| | - Sandrine Genetet
- Université Paris Cité and Université des Antilles, INSERM U1134, BIGR, F-75014 Paris, France
| | - Maria Florencia Leal Denis
- Instituto de Química y Fisico-Química Biológicas “Prof. Alejandro C. Paladini”, UBA, CONICET, Facultad de Farmacia y Bioquímica, 1113 Buenos Aires, Argentina
| | - Michael Dussiot
- Université Paris Cité, INSERM U1163, IMAGINE, F-75015 Paris, France
| | - Pablo Julio Schwarzbaum
- Instituto de Química y Fisico-Química Biológicas “Prof. Alejandro C. Paladini”, UBA, CONICET, Facultad de Farmacia y Bioquímica, 1113 Buenos Aires, Argentina
| | - Mariano Anibal Ostuni
- Université Paris Cité and Université des Antilles, INSERM U1134, BIGR, F-75014 Paris, France
| | - Isabelle Mouro-Chanteloup
- Université Paris Cité and Université des Antilles, INSERM U1134, BIGR, F-75014 Paris, France
- Correspondence:
| |
Collapse
|
17
|
Stomatin modulates adipogenesis through the ERK pathway and regulates fatty acid uptake and lipid droplet growth. Nat Commun 2022; 13:4174. [PMID: 35854007 PMCID: PMC9296665 DOI: 10.1038/s41467-022-31825-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 07/01/2022] [Indexed: 11/08/2022] Open
Abstract
Regulation of fatty acid uptake, lipid production and storage, and metabolism of lipid droplets (LDs), is closely related to lipid homeostasis, adipocyte hypertrophy and obesity. We report here that stomatin, a major constituent of lipid raft, participates in adipogenesis and adipocyte maturation by modulating related signaling pathways. In adipocyte-like cells, increased stomatin promotes LD growth or enlargements by facilitating LD-LD fusion. It also promotes fatty acid uptake from extracellular environment by recruiting effector molecules, such as FAT/CD36 translocase, to lipid rafts to promote internalization of fatty acids. Stomatin transgenic mice fed with high-fat diet exhibit obesity, insulin resistance and hepatic impairments; however, such phenotypes are not seen in transgenic animals fed with regular diet. Inhibitions of stomatin by gene knockdown or OB-1 inhibit adipogenic differentiation and LD growth through downregulation of PPARγ pathway. Effects of stomatin on PPARγ involves ERK signaling; however, an alternate pathway may also exist. Stomatin is a component of lipid rafts. Here, Wu et al. show that stomatin modulates the differentiation and functions of adipocytes by regulating adipogenesis signaling and fatty acid influx such that with excessive calorie intake, increased stomatin induces adiposity.
Collapse
|
18
|
Wu N, Zhu Y, Jiang W, Song Y, Yin L, Lu Y, Tao D, Liu Y, Ma Y. A novel NPHS2 mutation (c.865A > G) identified in a Chinese family with steroid-resistant nephrotic syndrome alters subcellular localization of nephrin. Genes Genomics 2022; 44:551-559. [PMID: 35099763 DOI: 10.1007/s13258-022-01220-5] [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: 11/01/2021] [Accepted: 01/16/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND NPHS2 is the causative gene of nephrotic syndrome type 2 (MIM 600995) which often clinically manifests as steroid-resistant nephrotic syndrome (SRNS). The NPHS2 gene encodes a slit diaphragm (SD) associated protein podocin. OBJECTIVE This study reported a novel disease-causing mutation of NPHS2 in a Chinese family with SRNS. We also investigated the pathogenic mechanism of the variants in this family. METHOD A Chinese family with SRNS was recruited. Whole exome sequencing was performed to screen for disease-causing mutation. Sanger sequencing was used to confirm the results. In vitro functional experiments including immunoblotting, co-immunoprecipitation and double immunofluorescence staining were performed to explore the pathogenic mechanisms of mutations. RESULTS In this family, compound heterozygous mutations of NPHS2 (c.467dupT and c.865A > G) were identified and segregated with the disease. The maternal c.865A > G was a novel variant, leading to amino acid substitution (p.K289E). In vitro functional assays indicated that c.467dupT (p.L156FfsX11) mutant lost interaction with nephrin. Both K289E and L156FfsX11 mutants showed sharply diminished plasma membrane localization. Furthermore, abnormal distribution of podocin mutants also altered the cell membrane localization of nephrin. CONCLUSION We reported a family with SRNS caused by compound heterozygous mutations of NPHS2 (c.467dupT and c.865A > G). c.865A > G (p.K289E) in NPHS2 was a novel causative variant associated with SRNS. Both variants in this family not only affected the normal cell membrane localization of podocin, but also altered the cell membrane localization of nephrin which is the major architectural protein of SD.
Collapse
Affiliation(s)
- Na Wu
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Yingchuan Zhu
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Wenhao Jiang
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Yue Song
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Lan Yin
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Yilu Lu
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Dachang Tao
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Yunqiang Liu
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China
| | - Yongxin Ma
- Department of Medical Genetics, West China Hospital, Sichuan University, 1st Keyuan 4 Lu, High-Tech Zone, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
19
|
Kataoka K, Suzuki S, Tenno T, Goda N, Hibino E, Oshima A, Hiroaki H. A cryptic phosphate-binding pocket on the SPFH domain of human stomatin that regulates a novel fibril-like self-assembly. Curr Res Struct Biol 2022; 4:158-166. [PMID: 35663930 PMCID: PMC9157467 DOI: 10.1016/j.crstbi.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/03/2022] Open
Abstract
Human stomatin (hSTOM) is a component of the membrane skeleton of erythrocytes that maintains the membrane's shape and stiffness through interconnecting spectrin and actin. hSTOM is a member of the protein family that possesses a single stomatin/prohibitin/flotillin/HflK (SPFH) domain at the center of the molecule. Although SPFH domain proteins are widely distributed from archaea to mammals, the detailed function of the domain remains unclear. In this study, we first determined the solution structure of the SPFH domain of hSTOM (hSTOM(SPFH)) via NMR. The solution structure of hSTOM(SPFH) is essentially identical to the already reported crystal structure of the STOM SPFH domain (mSTOM(SPFH)) of mice, except for the existence of a small hydrophilic pocket on the surface. We identified this pocket as a phosphate-binding site by comparing its NMR spectra with and without phosphate ions. Meanwhile, during the conventional process of protein NMR analysis, we eventually discovered that hSTOM(SPFH) formed a unique solid material after lyophilization. This lyophilized hSTOM(SPFH) sample was moderately slowly dissolved in a physiological buffer. Interestingly, it was resistant to dissolution against the phosphate buffer. We then found that the lyophilized hSTOM(SPFH) formed a fibril-like assembly under electron microscopy. Finally, we succeeded in reproducing this fibril-like assembly of hSTOM(SPFH) using a centrifugal ultrafiltration device, thus demonstrating that the increased protein concentration may promote self-assembly of hSTOM(SPFH) into fibril forms. Our observations may help understand the molecular function of the SPFH domain and its involvement in protein oligomerization as a component of the membrane skeleton. (245 words). Solution structure of human stomatin SPFH domain is determined. A cryptic phosphate-binding pocket was identified. Stomatin SPFH domain can form a fibril-like assembly at a high concentration. Phosphate ions promote formation of the fibril-like assembly.
Collapse
|
20
|
Structural organization of erythrocyte membrane microdomains and their relation with malaria susceptibility. Commun Biol 2021; 4:1375. [PMID: 34880413 PMCID: PMC8655059 DOI: 10.1038/s42003-021-02900-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
Cholesterol-rich microdomains are membrane compartments characterized by specific lipid and protein composition. These dynamic assemblies are involved in several biological processes, including infection by intracellular pathogens. This work provides a comprehensive analysis of the composition of human erythrocyte membrane microdomains. Based on their floating properties, we also categorized the microdomain-associated proteins into clusters. Interestingly, erythrocyte microdomains include the vast majority of the proteins known to be involved in invasion by the malaria parasite Plasmodium falciparum. We show here that the Ecto-ADP-ribosyltransferase 4 (ART4) and Aquaporin 1 (AQP1), found within one specific cluster, containing the essential host determinant CD55, are recruited to the site of parasite entry and then internalized to the newly formed parasitophorous vacuole membrane. By generating null erythroid cell lines, we showed that one of these proteins, ART4, plays a role in P. falciparum invasion. We also found that genetic variants in both ART4 and AQP1 are associated with susceptibility to the disease in a malaria-endemic population.
Collapse
|
21
|
Pushkarsky T, Ward A, Ivanov A, Lin X, Sviridov D, Nekhai S, Bukrinsky MI. Abundance of Nef and p-Tau217 in Brains of Individuals Diagnosed with HIV-Associated Neurocognitive Disorders Correlate with Disease Severance. Mol Neurobiol 2021; 59:1088-1097. [PMID: 34843091 PMCID: PMC8857174 DOI: 10.1007/s12035-021-02608-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/15/2021] [Indexed: 11/25/2022]
Abstract
HIV-associated neurocognitive disorders (HAND) is a term used to describe a variety of neurological impairments observed in HIV-infected individuals. The pathogenic mechanisms of HAND and of its connection to HIV infection remain unknown, but one of the considered hypotheses suggests that HIV infection accelerates the development of Alzheimer’s disease. Previous studies suggested that HIV-1 Nef may contribute to HAND by inhibiting cholesterol efflux, increasing the abundance of lipid rafts, and affecting their functionality. Our comparative analysis of postmortem brain samples demonstrated a trend toward the decreased abundance of cholesterol transporter ABCA1 in samples from HIV-infected ART-treated individuals relative to samples from uninfected controls, and a reverse correlation between ABCA1 and flotillin 1, a marker for lipid rafts, in all analyzed samples. The brain samples from HIV-infected individuals, both with and without HAND, were characterized by the increased abundance of p-Tau217 peptide, which correlated with the abundance of flotillin 1. HIV-1 Nef was analyzed in samples from HAND-affected individuals by Western blot with 4 different antibodies and by LC–MS/MS, producing a Nef-positivity score. A significant correlation was found between this score and the abundance of flotillin 1, the abundance of p-Tau217, and the severity of HAND. These results highlight the contribution of Nef and Nef-dependent impairment of cholesterol efflux to HAND pathogenesis and support a connection between the pathogenesis of HAND and Alzheimer’s disease.
Collapse
Affiliation(s)
- Tatiana Pushkarsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Adam Ward
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- The George Washington University Milken Institute School of Public Health, Washington, DC, USA
- Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Andrey Ivanov
- College of Medicine, Howard University, Washington, DC, USA
| | - Xionghao Lin
- College of Medicine, Howard University, Washington, DC, USA
- College of Dentistry, Howard University, Washington, DC, USA
| | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Sergei Nekhai
- College of Medicine, Howard University, Washington, DC, USA
| | - Michael I Bukrinsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
22
|
Scholz AS, Baur SSM, Wolf D, Bramkamp M. An Stomatin, Prohibitin, Flotillin, and HflK/C-Domain Protein Required to Link the Phage-Shock Protein to the Membrane in Bacillus subtilis. Front Microbiol 2021; 12:754924. [PMID: 34777311 PMCID: PMC8581546 DOI: 10.3389/fmicb.2021.754924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
Membrane surveillance and repair is of utmost importance to maintain cellular integrity and allow cellular life. Several systems detect cell envelope stress caused by antimicrobial compounds and abiotic stresses such as solvents, pH-changes and temperature in bacteria. Proteins containing an Stomatin, Prohibitin, Flotillin, and HflK/C (SPFH)-domain, including bacterial flotillins have been shown to be involved in membrane protection and membrane fluidity regulation. Here, we characterize a bacterial SPFH-domain protein, YdjI that is part of a stress induced complex in Bacillus subtilis. We show that YdjI is required to localize the ESCRT-III homolog PspA to the membrane with the help of two membrane integral proteins, YdjG/H. In contrast to classical flotillins, YdjI resides in fluid membrane regions and does not enrich in detergent resistant membrane fractions. However, similarly to FloA and FloT from B. subtilis, deletion of YdjI decreases membrane fluidity. Our data reveal a hardwired connection between phage shock response and SPFH proteins.
Collapse
Affiliation(s)
- Abigail Savietto Scholz
- Institute for General Microbiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sarah S. M. Baur
- Institute for General Microbiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Diana Wolf
- Institute of Microbiology, Technische Universität Dresden, Dresden, Germany
| | - Marc Bramkamp
- Institute for General Microbiology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
23
|
Barshtein G, Pajic-Lijakovic I, Gural A. Deformability of Stored Red Blood Cells. Front Physiol 2021; 12:722896. [PMID: 34690797 PMCID: PMC8530101 DOI: 10.3389/fphys.2021.722896] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022] Open
Abstract
Red blood cells (RBCs) deformability refers to the cells’ ability to adapt their shape to the dynamically changing flow conditions so as to minimize their resistance to flow. The high red cell deformability enables it to pass through small blood vessels and significantly determines erythrocyte survival. Under normal physiological states, the RBCs are attuned to allow for adequate blood flow. However, rigid erythrocytes can disrupt the perfusion of peripheral tissues and directly block microvessels. Therefore, RBC deformability has been recognized as a sensitive indicator of RBC functionality. The loss of deformability, which a change in the cell shape can cause, modification of cell membrane or a shift in cytosol composition, can occur due to various pathological conditions or as a part of normal RBC aging (in vitro or in vivo). However, despite extensive research, we still do not fully understand the processes leading to increased cell rigidity under cold storage conditions in a blood bank (in vitro aging), In the present review, we discuss publications that examined the effect of RBCs’ cold storage on their deformability and the biological mechanisms governing this change. We first discuss the change in the deformability of cells during their cold storage. After that, we consider storage-related alterations in RBCs features, which can lead to impaired cell deformation. Finally, we attempt to trace a causal relationship between the observed phenomena and offer recommendations for improving the functionality of stored cells.
Collapse
Affiliation(s)
- Gregory Barshtein
- Biochemistry Department, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Alexander Gural
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
24
|
Role of ERLINs in the Control of Cell Fate through Lipid Rafts. Cells 2021; 10:cells10092408. [PMID: 34572057 PMCID: PMC8470593 DOI: 10.3390/cells10092408] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022] Open
Abstract
ER lipid raft-associated protein 1 (ERLIN1) and 2 (ERLIN2) are 40 kDa transmembrane glycoproteins belonging to the family of prohibitins, containing a PHB domain. They are generally localized in the endoplasmic reticulum (ER), where ERLIN1 forms a heteroligomeric complex with its closely related ERLIN2. Well-defined functions of ERLINS are promotion of ER-associated protein degradation, mediation of inositol 1,4,5-trisphosphate (IP3) receptors, processing and regulation of lipid metabolism. Until now, ERLINs have been exclusively considered protein markers of ER lipid raft-like microdomains. However, under pathophysiological conditions, they have been described within mitochondria-associated endoplasmic reticulum membranes (MAMs), tethering sites between ER and mitochondria, characterized by the presence of specialized raft-like subdomains enriched in cholesterol and gangliosides, which play a key role in the membrane scrambling and function. In this context, it is emerging that ER lipid raft-like microdomains proteins, i.e., ERLINs, may drive mitochondria-ER crosstalk under both physiological and pathological conditions by association with MAMs, regulating the two main processes underlined, survival and death. In this review, we describe the role of ERLINs in determining cell fate by controlling the “interchange” between apoptosis and autophagy pathways, considering that their alteration has a significant impact on the pathogenesis of several human diseases.
Collapse
|
25
|
Koudatsu S, Masatani T, Konishi R, Asada M, Hakimi H, Kurokawa Y, Tomioku K, Kaneko O, Fujita A. Glycosphingolipid GM3 is localized in both exoplasmic and cytoplasmic leaflets of Plasmodium falciparum malaria parasite plasma membrane. Sci Rep 2021; 11:14890. [PMID: 34290278 PMCID: PMC8295280 DOI: 10.1038/s41598-021-94037-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
Lipid rafts, sterol-rich and sphingolipid-rich microdomains on the plasma membrane are important in processes like cell signaling, adhesion, and protein and lipid transport. The virulence of many eukaryotic parasites is related to raft microdomains on the cell membrane. In the malaria parasite Plasmodium falciparum, glycosylphosphatidylinositol-anchored proteins, which are important for invasion and are possible targets for vaccine development, are localized in the raft. However, rafts are poorly understood. We used quick-freezing and freeze-fracture immuno-electron microscopy to examine the localization of monosialotetrahexosylganglioside (GM1) and monosialodihexosylganglioside (GM3), putative raft microdomain components in P. falciparum and infected erythrocytes. This method immobilizes molecules in situ, minimizing artifacts. GM3 was localized in the exoplasmic (EF) and cytoplasmic leaflets (PF) of the parasite and the parasitophorous vacuole (PV) membranes, but solely in the EF of the infected erythrocyte membrane, as in the case for uninfected erythrocytes. Phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2) was localized solely in the PF of erythrocyte, parasite, and PV membranes. This is the first time that GM3, the major component of raft microdomains, was found in the PF of a biological membrane. The unique localization of raft microdomains may be due to P. falciparum lipid metabolism and its unique biological processes, like protein transport from the parasite to infected erythrocytes.
Collapse
Affiliation(s)
- Shiomi Koudatsu
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan
| | - Tatsunori Masatani
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan.,Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Rikako Konishi
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan
| | - Masahito Asada
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan.,National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, 080-8555, Japan
| | - Hassan Hakimi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan.,National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, 080-8555, Japan
| | - Yuna Kurokawa
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan
| | - Kanna Tomioku
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Akikazu Fujita
- Department of Molecular and Cell Biology and Biochemistry, Basic Veterinary Science, Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima, 890-0065, Japan.
| |
Collapse
|
26
|
A new role of glutathione peroxidase 4 during human erythroblast enucleation. Blood Adv 2021; 4:5666-5680. [PMID: 33211827 DOI: 10.1182/bloodadvances.2020003100] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The selenoprotein glutathione peroxidase 4 (GPX4), the only member of the glutathione peroxidase family able to directly reduce cell membrane-oxidized fatty acids and cholesterol, was recently identified as the central regulator of ferroptosis. GPX4 knockdown in mouse hematopoietic cells leads to hemolytic anemia and to increased spleen erythroid progenitor death. The role of GPX4 during human erythropoiesis is unknown. Using in vitro erythroid differentiation, we show here that GPX4-irreversible inhibition by 1S,3R-RSL3 (RSL3) and its short hairpin RNA-mediated knockdown strongly impaired enucleation in a ferroptosis-independent manner not restored by tocopherol or iron chelators. During enucleation, GPX4 localized with lipid rafts at the cleavage furrows between reticulocytes and pyrenocytes. Its inhibition impacted enucleation after nuclear condensation and polarization and was associated with a defect in lipid raft clustering (cholera toxin staining) and myosin-regulatory light-chain phosphorylation. Because selenoprotein translation and cholesterol synthesis share a common precursor, we investigated whether the enucleation defect could represent a compensatory mechanism favoring GPX4 synthesis at the expense of cholesterol, known to be abundant in lipid rafts. Lipidomics and filipin staining failed to show any quantitative difference in cholesterol content after RSL3 exposure. However, addition of cholesterol increased cholera toxin staining and myosin-regulatory light-chain phosphorylation, and improved enucleation despite GPX4 knockdown. In summary, we identified GPX4 as a new actor of human erythroid enucleation, independent of its function in ferroptosis control. We described its involvement in lipid raft organization required for contractile ring assembly and cytokinesis, leading in fine to nucleus extrusion.
Collapse
|
27
|
Tobys D, Kowalski LM, Cziudaj E, Müller S, Zentis P, Pach E, Zigrino P, Blaeske T, Höning S. Inhibition of clathrin-mediated endocytosis by knockdown of AP-2 leads to alterations in the plasma membrane proteome. Traffic 2020; 22:6-22. [PMID: 33225555 DOI: 10.1111/tra.12770] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023]
Abstract
In eukaryotic cells, clathrin-mediated endocytosis (CME) is a central pathway for the internalization of proteins from the cell surface, thereby contributing to the maintenance of the plasma membrane protein composition. A key component for the formation of endocytic clathrin-coated vesicles (CCVs) is AP-2, as it sequesters cargo membrane proteins, recruits a multitude of other endocytic factors and initiates clathrin polymerization. Here, we inhibited CME by depletion of AP-2 and explored the consequences for the plasma membrane proteome. Quantitative analysis revealed accumulation of major constituents of the endosomal-lysosomal system reflecting a block in retrieval by compensatory CME. The noticeable enrichment of integrins and blockage of their turnover resulted in severely impaired cell migration. Rare proteins such as the anti-cancer drug target CA9 and tumor markers (CD73, CD164, CD302) were significantly enriched. The AP-2 knockdown attenuated the global endocytic capacity, but clathrin-independent entry pathways were still operating, as indicated by persistent internalization of specific membrane-spanning and GPI-anchored receptors (PVR, IGF1R, CD55, TNAP). We hypothesize that blocking AP-2 function and thus inhibiting CME may be a novel approach to identify new druggable targets, or to increase their residence time at the plasma membrane, thereby increasing the probability for efficient therapeutic intervention.
Collapse
Affiliation(s)
- David Tobys
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Lisa Maria Kowalski
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Eva Cziudaj
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Stefan Müller
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Peter Zentis
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Elke Pach
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tobias Blaeske
- Department of Plant Physiology and Biochemistry, University of Constance, Constance, Germany
| | - Stefan Höning
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
28
|
Yang W, Geng C, Yang Z, Xu B, Shi W, Yang Y, Tian Y. Deciphering the roles of caveolin in neurodegenerative diseases: The good, the bad and the importance of context. Ageing Res Rev 2020; 62:101116. [PMID: 32554058 DOI: 10.1016/j.arr.2020.101116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases (NDDs), which contribute to progressive and irreversible impairments of both the structure and function of the nervous system, pose a substantial socioeconomic burden on society. Mitochondrial dysfunction, oxidative stress, membrane damage, DNA damage, and abnormal protein degradation pathways play pivotal roles in the etiology of NDDs. Recently, growing evidence has demonstrated that caveolins are important in the pathology of NDDs due to their cellular functions in signal transduction, endocytosis, transcytosis, cholesterol transport, and lipid homeostasis. Given the significance of caveolins, here we review the literature to clarify their molecular mechanisms and roles in NDDs. We first briefly introduce the general background on caveolins. Next, we focus on the various important functions of caveolins in the brain. Finally, we emphasize recent progress regarding caveolins, especially Cav-1, which exert both benefit and unfavorable effects in NDDs such as AD and PD. Collectively, the data presented here should advance the investigation of caveolins for the future development of innovative strategies for the treatment of NDDs.
Collapse
Affiliation(s)
- Wenwen Yang
- Department of Medical Research Center, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, 10 Fengcheng Three Road, Xi'an 710021, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chenhui Geng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Zhi Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Baoping Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Wenzhen Shi
- Department of Medical Research Center, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, 10 Fengcheng Three Road, Xi'an 710021, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Life of Sciences, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| | - Ye Tian
- Department of Medical Research Center, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, 10 Fengcheng Three Road, Xi'an 710021, China.
| |
Collapse
|
29
|
Zhang X, Bandyopadhyay S, Araujo LP, Tong K, Flores J, Laubitz D, Zhao Y, Yap G, Wang J, Zou Q, Ferraris R, Zhang L, Hu W, Bonder EM, Kiela PR, Coffey R, Verzi MP, Ivanov II, Gao N. Elevating EGFR-MAPK program by a nonconventional Cdc42 enhances intestinal epithelial survival and regeneration. JCI Insight 2020; 5:135923. [PMID: 32686657 PMCID: PMC7455142 DOI: 10.1172/jci.insight.135923] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/09/2020] [Indexed: 01/05/2023] Open
Abstract
The regulatory mechanisms enabling the intestinal epithelium to maintain a high degree of regenerative capacity during mucosal injury remain unclear. Ex vivo survival and clonogenicity of intestinal stem cells (ISCs) strictly required growth response mediated by cell division control 42 (Cdc42) and Cdc42-deficient enteroids to undergo rapid apoptosis. Mechanistically, Cdc42 engaging with EGFR was required for EGF-stimulated, receptor-mediated endocytosis and sufficient to promote MAPK signaling. Proteomics and kinase analysis revealed that a physiologically, but nonconventionally, spliced Cdc42 variant 2 (V2) exhibited stronger MAPK-activating capability. Human CDC42-V2 is transcriptionally elevated in some colon tumor tissues. Accordingly, mice engineered to overexpress Cdc42-V2 in intestinal epithelium showed elevated MAPK signaling, enhanced regeneration, and reduced mucosal damage in response to irradiation. Overproducing Cdc42-V2 specifically in mouse ISCs enhanced intestinal regeneration following injury. Thus, the intrinsic Cdc42-MAPK program is required for intestinal epithelial regeneration, and elevating this signaling cascade is capable of initiating protection from genotoxic injury.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
| | - Sheila Bandyopadhyay
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
| | - Leandro Pires Araujo
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Kevin Tong
- Department of Genetics, Division of Life Sciences, School of Arts and Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Juan Flores
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
| | - Daniel Laubitz
- Department of Pediatrics, University of Arizona, Tucson, Arizona, USA
| | - Yanlin Zhao
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - George Yap
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jingren Wang
- Department of Mechanical and Aerospace Engineering, School of Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Qingze Zou
- Department of Mechanical and Aerospace Engineering, School of Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Ronaldo Ferraris
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Lanjing Zhang
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
- Department of Pathology, University Medical Center of Princeton, Plainsboro, New Jersey, USA
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Edward M. Bonder
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
| | - Pawel R. Kiela
- Department of Pediatrics, University of Arizona, Tucson, Arizona, USA
| | - Robert Coffey
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, and Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael P. Verzi
- Department of Genetics, Division of Life Sciences, School of Arts and Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Ivaylo I. Ivanov
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Nan Gao
- Department of Biological Sciences, Division of Life Sciences, School of Arts and Sciences, Rutgers University, Newark, New Jersey, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
30
|
McVey MJ, Kuebler WM, Orbach A, Arbell D, Zelig O, Barshtein G, Yedgar S. Reduced deformability of stored red blood cells is associated with generation of extracellular vesicles. Transfus Apher Sci 2020; 59:102851. [PMID: 32571640 DOI: 10.1016/j.transci.2020.102851] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/12/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Throughout storage, red blood cells (RBCs) undergo detrimental changes in viability and their ability to effectively transport oxygen. RBC storage lesions are mediated, in part, by a progressive loss of cell deformability, and associated with the release of extracellular vesicles (EVs). Accumulation of EVs during the storage of RBCs correlates with a decrease in RBC surface area to volume ratio. Similarly, the loss of RBC-deformability is associated with loss of RBC surface area to volume ratio. In this study we thus tested whether loss of RBC-deformability is associated with increased RBC-EV production during blood storage. EVs obtained by differential centrifugation of stored RBCs (non-leukoreduced non-irradiated or leukoreduced γ-irradiated RBCs stored 35 or 28 days respectively) were enumerated by high-sensitivity flow cytometry. RBC deformability was quantified, using a cell-flow-properties-analyzer, by measuring the median cell elongation ratio (MER) and percentage of low and high deformable cells in the population (%, LDFC, and HDFC, respectively). The number of EVs was inversely correlated with the MER and positively correlated with the %LDFC with both measures showing highly significant logarithmic dependence with EV levels in stored RBCs. Considering how highly deformable cells did not correlate with EV formation as compared with low deformable RBCs we propose that the formation of EVs is a key factor leading to increased RBC-rigidity.
Collapse
Affiliation(s)
- M J McVey
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, United States; Departments of Physiology and Anesthesia, University of Toronto, Toronto, ON, United States; Hospital for Sick Children, Department of Anesthesia and Pain Medicine, United States; Department of Physics, Ryerson University, Toronto, ON, United States
| | - W M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, United States; Departments of Physiology and Surgery, University of Toronto, Toronto, ON, United States; Institute of Physiology, Charité-Universitätsmedizin Berlin, Germany
| | - A Orbach
- Department of Biochemistry, Hebrew University, Faculty of Medicine, Jerusalem, Israel
| | - D Arbell
- Department of Pediatric Surgery, Hadassah- Hebrew University Hospital, Jerusalem, Israel
| | - O Zelig
- Blood Bank, Hadassah University Hospital, Jerusalem, Israel
| | - G Barshtein
- Department of Biochemistry, Hebrew University, Faculty of Medicine, Jerusalem, Israel.
| | - S Yedgar
- Department of Biochemistry, Hebrew University, Faculty of Medicine, Jerusalem, Israel
| |
Collapse
|
31
|
Skryabin GO, Komelkov AV, Savelyeva EE, Tchevkina EM. Lipid Rafts in Exosome Biogenesis. BIOCHEMISTRY (MOSCOW) 2020; 85:177-191. [PMID: 32093594 DOI: 10.1134/s0006297920020054] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Exosomes (secreted extracellular vesicles formed in the intracellular vesicular transport system) play a crucial role in distant cell-cell communication. Exosomes transfer active forms of various biomolecules; the molecular composition of the exosomal cargo is a result of targeted selection and depends on the type of producer cells. The mechanisms underlying exosome formation and cargo selection are poorly understood. It is believed that there are several pathways for exosome biogenesis, although the questions about their independence and simultaneous coexistence in the cell still remain open. The least studied topic is the recently discovered mechanism of exosome formation associated with lipid rafts, or membrane lipid microdomains. Here, we present modern concepts and basic hypotheses on the mechanisms of exosome biogenesis and secretion and summarize current data on the involvement of lipid rafts and their constituent molecules in these processes. Special attention is paid to the analysis of possible role in the exosome formation of raft-forming proteins of the SPFH family, components of planar rafts, and caveolin, the main component of caveolae.
Collapse
Affiliation(s)
- G O Skryabin
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - A V Komelkov
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia.
| | - E E Savelyeva
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| | - E M Tchevkina
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115478, Russia
| |
Collapse
|
32
|
Jin Y, Liang Q, Tieleman DP. Interactions between Band 3 Anion Exchanger and Lipid Nanodomains in Ternary Lipid Bilayers: Atomistic Simulations. J Phys Chem B 2020; 124:3054-3064. [DOI: 10.1021/acs.jpcb.0c01055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yapan Jin
- Center for Statistical and Theoretical Condensed Matter Physics and Department of Physics, Zhejiang Normal University, Jinhua 321004, P. R. China
| | - Qing Liang
- Center for Statistical and Theoretical Condensed Matter Physics and Department of Physics, Zhejiang Normal University, Jinhua 321004, P. R. China
| | - D. Peter Tieleman
- Centre for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
33
|
Flotillins: At the Intersection of Protein S-Palmitoylation and Lipid-Mediated Signaling. Int J Mol Sci 2020; 21:ijms21072283. [PMID: 32225034 PMCID: PMC7177705 DOI: 10.3390/ijms21072283] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Flotillin-1 and flotillin-2 are ubiquitously expressed, membrane-associated proteins involved in multifarious cellular events from cell signaling, endocytosis, and protein trafficking to gene expression. They also contribute to oncogenic signaling. Flotillins bind the cytosolic leaflet of the plasma membrane and endomembranes and, upon hetero-oligomerization, serve as scaffolds facilitating the assembly of multiprotein complexes at the membrane-cytosol interface. Additional functions unique to flotillin-1 have been discovered recently. The membrane-binding of flotillins is regulated by S-palmitoylation and N-myristoylation, hydrophobic interactions involving specific regions of the polypeptide chain and, to some extent, also by their oligomerization. All these factors endow flotillins with an ability to associate with the sphingolipid/cholesterol-rich plasma membrane domains called rafts. In this review, we focus on the critical input of lipids to the regulation of the flotillin association with rafts and thereby to their functioning. In particular, we discuss how the recent developments in the field of protein S-palmitoylation have contributed to the understanding of flotillin1/2-mediated processes, including endocytosis, and of those dependent exclusively on flotillin-1. We also emphasize that flotillins affect directly or indirectly the cellular levels of lipids involved in diverse signaling cascades, including sphingosine-1-phosphate and PI(4,5)P2. The mutual relations between flotillins and distinct lipids are key to the regulation of their involvement in numerous cellular processes.
Collapse
|
34
|
Minetti G, Bernecker C, Dorn I, Achilli C, Bernuzzi S, Perotti C, Ciana A. Membrane Rearrangements in the Maturation of Circulating Human Reticulocytes. Front Physiol 2020; 11:215. [PMID: 32256383 PMCID: PMC7092714 DOI: 10.3389/fphys.2020.00215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
Red blood cells (RBCs) begin their circulatory life as reticulocytes (Retics) after their egress from the bone marrow where, as R1 Retics, they undergo significant rearrangements in their membrane and intracellular components, via autophagic, proteolytic, and vesicle-based mechanisms. Circulating, R2 Retics must complete this maturational process, which involves additional loss of significant amounts of membrane and selected membrane proteins. Little is known about the mechanism(s) at the basis of this terminal differentiation in the circulation, which culminates with the production of a stable biconcave discocyte. The membrane of R1 Retics undergoes a selective remodeling through the release of exosomes that are enriched in transferrin receptor and membrane raft proteins and lipids, but are devoid of Band 3, glycophorin A, and membrane skeletal proteins. We wondered whether a similar selective remodeling occurred also in the maturation of R2 Retics. Peripheral blood R2 Retics, isolated by an immunomagnetic method, were compared with mature circulating RBCs from the same donor and their membrane protein and lipid content was analyzed. Results show that both Band 3 and spectrin decrease from R2 Retics to RBCs on a "per cell" basis. Looking at membrane proteins that are considered as markers of membrane rafts, flotillin-2 appears to decrease in a disproportionate manner with respect to Band 3. Stomatin also decreases but in a more proportionate manner with respect to Band 3, hinting at a heterogeneous nature of membrane rafts. High resolution lipidomics analysis, on the contrary, revealed that those lipids that are typically representative of the membrane raft phase, sphingomyelin and cholesterol, are enriched in mature RBCs with respct to Retics, relative to total cell lipids, strongly arguing in favor of the selective retention of at least certain subclasses of membrane rafts in RBCs as they mature from Retics. Our hypothesis that rafts serve as additional anchoring sites for the lipid bilayer to the underlying membrane-skeleton is corroborated by the present results. It is becoming ever more clear that a proper lipid composition of the reticulocyte is necessary for the production of a normal mature RBC.
Collapse
Affiliation(s)
- Giampaolo Minetti
- Laboratories of Biochemistry, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Claudia Bernecker
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Isabel Dorn
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Cesare Achilli
- Laboratories of Biochemistry, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Stefano Bernuzzi
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Cesare Perotti
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Annarita Ciana
- Laboratories of Biochemistry, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
35
|
Yokoyama H, Matsui I. The lipid raft markers stomatin, prohibitin, flotillin, and HflK/C (SPFH)-domain proteins form an operon with NfeD proteins and function with apolar polyisoprenoid lipids. Crit Rev Microbiol 2020; 46:38-48. [PMID: 31983249 DOI: 10.1080/1040841x.2020.1716682] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SPFH-domain proteins are found in almost all organisms across three domains: archaea, bacteria, and eukaryotes. In eukaryotic organelles, their subfamilies exhibit overlapping distribution and functions; thus, the rationality of annotation to discriminate these subfamilies remains unclear. In this review, the binding ability of prokaryotic SPFH-domain proteins towards nonpolar polyisoprenoides such as squalene and lycopene, rather than cholesterol, is discussed. The hydrophobic region at the C-terminus of SPFH-domain proteins constitutes the main region that binds apolar polyisoprenoid lipids as well as cholesterol and substantively contributes towards lipid raft formation as these regions are self-assembled together with specific lipids. Because the scaffolding proteins caveolins show common topological properties with SPFH-domain proteins such as stomatin and flotillin, the α-helical segments of stomatin proteins can flexibly move along with the membrane surface, with such movement potentially leading to membrane bending via lipid raft clustering through the formation of high order homo-oligomeric complexes of SPFH-domain proteins. We also discuss the functional significance and ancient origin of SPFH-domain proteins and the NfeD protein (STOPP) operon, which can be traced back to the ancient living cells that diverged and evolved to archaea and bacteria. Based on the molecular mechanism whereby the STOPP-protease degrades the C-terminal hydrophobic clusters of SPFH-domain proteins, it is conceivable that STOPP-protease might control the physicochemical properties of lipid rafts.
Collapse
Affiliation(s)
- Hideshi Yokoyama
- Department of Medical and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Ikuo Matsui
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
36
|
Hornung TC, Biesalski HK. Glut-1 explains the evolutionary advantage of the loss of endogenous vitamin C-synthesis: The electron transfer hypothesis. EVOLUTION MEDICINE AND PUBLIC HEALTH 2019; 2019:221-231. [PMID: 31857900 PMCID: PMC6915226 DOI: 10.1093/emph/eoz024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 07/30/2019] [Indexed: 12/27/2022]
Abstract
Introduction During evolution, some species including humans, monkeys and fruit bats lost the ability for ascorbic acid (AA) biosynthesis due to inactivation of the enzyme l-gulono-lactone oxidase (GLO) and subsequently became dependent on dietary vitamin C. There are four current hypotheses in relation to the benefit of vitamin C dependence in the context of adaptation and reproduction. Here we advance and test a new ‘electron transfer hypothesis’, which focusses on the role of the expression of glucose transporter 1 (Glut-1) in red blood cells (RBCs) in recycling vitamin C, thereby increasing the efficiency of micronutrient uptake. Methods To evaluate the benefit of Glut-1 expression, we determined vitamin C uptake into RBCs and potential release from two different species, humans with l-Gulono-lactone-oxidase (GLO-loss) and pigs with functional GLO. Results The oxidized form of vitamin C (dehydroascorbate, DHA) was transported into human RBCs via Glut-1. There was no transport of either the reduced (AA) or the oxidized vitamin in pig erythrocytes. Conclusion We propose that the transport of vitamin C increases an intracellular electron pool, which transfers electrons from intracellular ascorbate to extracellular substances like ascorbyl free radical or DHA, resulting in 100-fold smaller daily requirement of this essential redox sensitive micronutrient. This would be an advantage during seasonal changes of the availability from food and may be the key for the survival of individuals without vitamin C biosynthesis. Lay Summary 40 million years ago some individuals lost the ability to synthesize vitamin C. Why did they survive such as humans until now? Individuals with a specific glucose transporter Glut-1 on their erythrocytes which transports vitamin C need less and are protected from scarcity due to seasons and food competitors.
Collapse
Affiliation(s)
- Tabea C Hornung
- Department of Nutrition, University of Hohenheim, Garbenstrasse 30, Stuttgart 70593, Germany
| | - Hans-Konrad Biesalski
- Department of Nutrition, University of Hohenheim, Garbenstrasse 30, Stuttgart 70593, Germany
| |
Collapse
|
37
|
Thalwieser Z, Király N, Fonódi M, Csortos C, Boratkó A. Protein phosphatase 2A-mediated flotillin-1 dephosphorylation up-regulates endothelial cell migration and angiogenesis regulation. J Biol Chem 2019; 294:20196-20206. [PMID: 31753918 DOI: 10.1074/jbc.ra119.007980] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells have key functions in endothelial barrier integrity and in responses to angiogenic signals that promote cell proliferation, cell migration, cytoskeletal reorganization, and formation of new blood vessels. These functions highly depend on protein-protein interactions in cell-cell junction and cell attachment complexes and on interactions with cytoskeletal proteins. Protein phosphatase 2A (PP2A) dephosphorylates several target proteins involved in cytoskeletal dynamics and cell adhesion. Our goal was to find new interacting and substrate proteins of the PP2A-B55α holoenzyme in bovine pulmonary endothelial cells. Using LC-MS/MS analysis, we identified flotillin-1 as a protein that binds recombinant GSH S-transferase-tagged PP2A-B55α. Immunoprecipitation experiments, proximity ligation assays, and immunofluorescent staining confirmed the interaction between these two endogenous proteins in endothelial cells. Originally, flotillins were described as regulatory proteins for axon regeneration, but they appear to function in many cellular processes, such as membrane receptor signaling, endocytosis, and cell adhesion. Ser315 is a known PKC-targeted site in flotillin-1. Utilizing phosphomutants of flotillin-1 and the NanoBiT luciferase assay, we show here that phosphorylation/dephosphorylation of Ser315 in flotillin-1 significantly affects its interaction with PP2A-B55α and that PP2A-B55α dephosphorylates phospho-Ser315 Spreading, attachment, migration, and in vitro tube formation rates of S315A variant-overexpressing cells were faster than those of nontransfected or S315D-transfected cells. These results indicate that the PP2A-flotillin-1 interaction identified here affects major physiological activities of pulmonary endothelial cells.
Collapse
Affiliation(s)
- Zsófia Thalwieser
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Nikolett Király
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Márton Fonódi
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Csilla Csortos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| |
Collapse
|
38
|
Vidal M. Exosomes: Revisiting their role as "garbage bags". Traffic 2019; 20:815-828. [PMID: 31418976 DOI: 10.1111/tra.12687] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
In recent years, the term "extracellular vesicle" (EV) has been used to define different types of vesicles released by various cells. It includes plasma membrane-derived vesicles (ectosomes/microvesicles) and endosome-derived vesicles (exosomes). Although it remains difficult to evaluate the compartment of origin of the two kinds of vesicles once released, it is critical to discriminate these vesicles because their mode of biogenesis is probably directly related to their physiologic function and/or to the physio-pathologic state of the producing cell. The purpose of this review is to specifically consider exosome secretion and its consequences in terms of a material loss for producing cells, rather than on the effects of exosomes once they are taken up by recipient cells. I especially describe one putative basic function of exosomes, that is, to convey material out of cells for off-site degradation by recipient cells. As illustrated by some examples, these components could be evacuated from cells for various reasons, for example, to promote "differentiation" or enhance homeostatic responses. This basic function might explain why so many diseases have made use of the exosomal pathway during pathogenesis.
Collapse
Affiliation(s)
- Michel Vidal
- LPHI - Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
39
|
Houck KL, Yuan H, Tian Y, Solomon M, Cramer D, Liu K, Zhou Z, Wu X, Zhang J, Oehler V, Dong JF. Physical proximity and functional cooperation of glycoprotein 130 and glycoprotein VI in platelet membrane lipid rafts. J Thromb Haemost 2019; 17:1500-1510. [PMID: 31145836 DOI: 10.1111/jth.14525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/28/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Clinical and laboratory studies have demonstrated that platelets become hyperactive and prothrombotic in conditions of inflammation. We have previously shown that the proinflammatory cytokine interleukin (IL)-6 forms a complex with soluble IL-6 receptor α (sIL-6Rα) to prime platelets for activation by subthreshold concentrations of collagen. Upon being stimulated with collagen, the transcription factor signal transducer and activator of transcription (STAT) 3 in platelets is phosphorylated and dimerized to act as a protein scaffold to facilitate the catalytic action between the kinase Syk and the substrate phospholipase Cγ2 (PLCγ2) in collagen-induced signaling. However, it remains unknown how collagen induces phosphorylation and dimerization of STAT3. METHODS AND RESULTS We conducted complementary in vitro experiments to show that the IL-6 receptor subunit glycoprotein 130 (GP130) was in physical proximity to the collagen receptor glycoprotein VI (GPVI in membrane lipid rafts of platelets. This proximity allows collagen to induce STAT3 activation and dimerization, and the IL-6-sIL-6Rα complex to activate the kinase Syk and the substrate PLCγ2 in the GPVI signal pathway, resulting in an enhanced platelet response to collagen. Disrupting lipid rafts or blocking GP130-Janus tyrosine kinase (JAK)-STAT3 signaling abolished the cross-activation and reduced platelet reactivity to collagen. CONCLUSION These results demonstrate cross-talk between collagen and IL-6 signal pathways. This cross-talk could potentially provide a novel mechanism for inflammation-induced platelet hyperactivity, so the IL-6-GP130-JAK-STAT3 pathway has been identified as a potential target to block this hyperactivity.
Collapse
Affiliation(s)
| | - Hengjie Yuan
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ye Tian
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | | | - Drake Cramer
- Bloodworks Research Institute, Seattle, Washington
| | - Kitty Liu
- Bloodworks Research Institute, Seattle, Washington
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
| | - Xiaoping Wu
- Bloodworks Research Institute, Seattle, Washington
| | - Jianning Zhang
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | - Vivian Oehler
- Clinical Research Division, Hutchison Cancer Center, Seattle, Washington
- Seattle Cancer Alliances, Seattle, Washington
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, Washington
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
40
|
Saito M, Cui L, Hirano M, Li G, Yanagisawa T, Sato T, Sukegawa J. Activity of Adenylyl Cyclase Type 6 Is Suppressed by Direct Binding of the Cytoskeletal Protein 4.1G. Mol Pharmacol 2019; 96:441-451. [PMID: 31383768 DOI: 10.1124/mol.119.116426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022] Open
Abstract
The G protein-coupled receptor (GPCR) signaling pathways mediated by trimeric G proteins have been extensively elucidated, but their associated regulatory mechanisms remain unclear. Parathyroid hormone (PTH)/PTH-related protein receptor (PTHR) is a GPCR coupled with Gs and Gq Gs activates adenylyl cyclases (ACs), which produces cAMP to regulate various cell fates. We previously showed that cell surface expression of PTHR was increased by its direct interaction with a subcortical cytoskeletal protein, 4.1G, whereas PTHR-mediated Gs/AC/cAMP signaling was suppressed by 4.1G through an unknown mechanism in human embryonic kidney (HEK)293 cells. In the present study, we found that AC type 6 (AC6), one of the major ACs activated downstream of PTHR, interacts with 4.1G in HEK293 cells, and the N-terminus of AC6 (AC6-N) directly and selectively binds to the 4.1/ezrin/radixin/moesin (FERM) domain of 4.1G (4.1G-FERM) in vitro. AC6-N was distributed at the plasma membrane, which was disturbed by knockdown of 4.1G. An AC6-N mutant, AC6-N-3A, in which three consecutive arginine residues are mutated to alanine residues, altered both binding to 4.1G-FERM and its plasma membrane distribution in vivo. Further, we overexpressed AC6-N to competitively inhibit the interaction of endogenous AC6 and 4.1G in cells. cAMP production induced by forskolin, an adenylyl cyclase activator, and PTH-(1-34) was enhanced by AC6-N expression and 4.1G-knockdown. In contrast, AC6-N-3A had no impact on forskolin- and PTH-(1-34)-induced cAMP productions. These data provide a novel regulatory mechanism that AC6 activity is suppressed by the direct binding of 4.1G to AC6-N, resulting in attenuation of PTHR-mediated Gs/AC6/cAMP signaling.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Linran Cui
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Marina Hirano
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Guanjie Li
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Teruyuki Yanagisawa
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Takeya Sato
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| | - Jun Sukegawa
- Department of Molecular Pharmacology, Tohoku University School of Medicine, Sendai, Miyagi, Japan (M.S., L.C., M.H., G.L., T.Y., T.S., J.S.); Department of Human Health and Nutrition, Shokei Gakuin University, Natori, Miyagi, Japan (M.H., J.S.); and Faculty of Health Sciences, Tohoku Fukushi University, Sendai, Miyagi, Japan (T.Y.)
| |
Collapse
|
41
|
Takano R, Kozuka-Hata H, Kondoh D, Bochimoto H, Oyama M, Kato K. A High-Resolution Map of SBP1 Interactomes in Plasmodium falciparum-infected Erythrocytes. iScience 2019; 19:703-714. [PMID: 31476617 PMCID: PMC6728614 DOI: 10.1016/j.isci.2019.07.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/09/2019] [Accepted: 07/19/2019] [Indexed: 11/19/2022] Open
Abstract
The pathogenesis of malaria parasites depends on host erythrocyte modifications that are facilitated by parasite proteins exported to the host cytoplasm. These exported proteins form a trafficking complex in the host cytoplasm that transports virulence determinants to the erythrocyte surface; this complex is thus essential for malaria virulence. Here, we report a comprehensive interaction network map of this complex. We developed authentic, unbiased, highly sensitive proteomic approaches to determine the proteins that interact with a core component of the complex, SBP1 (skeleton-binding protein 1). SBP1 interactomes revealed numerous exported proteins and potential interactors associated with SBP1 intracellular trafficking. We identified several host-parasite protein interactions and linked the exported protein MAL8P1.4 to Plasmodium falciparum virulence in infected erythrocytes. Our study highlights the complicated interplay between parasite and host proteins in the host cytoplasm and provides an interaction dataset connecting dozens of exported proteins required for P. falciparum virulence. We used shotgun proteomics to identify SBP1-interacting factors System validation showed complex interplay between parasite and host proteins Our system can be used to explore protozoan parasite virulence in erythrocytes
Collapse
Affiliation(s)
- Ryo Takano
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Hiroko Kozuka-Hata
- Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Daisuke Kondoh
- Laboratory of Veterinary Anatomy, Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Hiroki Bochimoto
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Masaaki Oyama
- Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Kentaro Kato
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan; Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Naruko-onsen, Osaki, Miyagi 989-6711, Japan.
| |
Collapse
|
42
|
Freitas Leal JK, Preijers F, Brock R, Adjobo-Hermans M, Bosman G. Red Blood Cell Homeostasis and Altered Vesicle Formation in Patients With Paroxysmal Nocturnal Hemoglobinuria. Front Physiol 2019; 10:578. [PMID: 31156458 PMCID: PMC6529780 DOI: 10.3389/fphys.2019.00578] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
A subset of the red blood cells (RBCs) of patients with paroxysmal nocturnal hemoglobinuria (PNH) lacks GPI-anchored proteins. Some of these proteins, such as CD59, inhibit complement activation and protect against complement-mediated lysis. This pathology thus provides the possibility to explore the involvement of complement in red blood cell homeostasis and the role of GPI-anchored proteins in the generation of microvesicles (MVs) in vivo. Detailed analysis of morphology, volume, and density of red blood cells with various CD59 expression levels from patients with PNH did not provide indications for a major aberration of the red blood cell aging process in patients with PNH. However, our data indicate that the absence of GPI-anchored membrane proteins affects the composition of red blood cell-derived microvesicles, as well as the composition and concentration of platelet-derived vesicles. These data open the way toward a better understanding on the pathophysiological mechanism of PNH and thereby to the development of new treatment strategies.
Collapse
Affiliation(s)
| | - Frank Preijers
- Laboratory for Hematology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Roland Brock
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, Netherlands
| | - Merel Adjobo-Hermans
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, Netherlands
| | - Giel Bosman
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
43
|
Rivera A, Vandorpe DH, Shmukler BE, Andolfo I, Iolascon A, Archer NM, Shabani E, Auerbach M, Hamerschlak N, Morton J, Wohlgemuth JG, Brugnara C, Snyder LM, Alper SL. Erythrocyte ion content and dehydration modulate maximal Gardos channel activity in KCNN4 V282M/+ hereditary xerocytosis red cells. Am J Physiol Cell Physiol 2019; 317:C287-C302. [PMID: 31091145 DOI: 10.1152/ajpcell.00074.2019] [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] [Indexed: 12/12/2022]
Abstract
Hereditary xerocytosis (HX) is caused by missense mutations in either the mechanosensitive cation channel PIEZO1 or the Ca2+-activated K+ channel KCNN4. All HX-associated KCNN4 mutants studied to date have revealed increased current magnitude and red cell dehydration. Baseline KCNN4 activity was increased in HX red cells heterozygous for KCNN4 mutant V282M. However, HX red cells maximally stimulated by Ca2+ ionophore A23187 or by PMCA Ca2+-ATPase inhibitor orthovanadate displayed paradoxically reduced KCNN4 activity. This reduced Ca2+-stimulated mutant KCNN4 activity in HX red cells was associated with unchanged sensitivity to KCNN4 inhibitor senicapoc and KCNN4 activator Ca2+, with slightly elevated Ca2+ uptake and reduced PMCA activity, and with decreased KCNN4 activation by calpain inhibitor PD150606. The altered intracellular monovalent cation content of HX red cells prompted experimental nystatin manipulation of red cell Na and K contents. Nystatin-mediated reduction of intracellular K+ with corresponding increase in intracellular Na+ in wild-type cells to mimic conditions of HX greatly suppressed vanadate-stimulated and A23187-stimulated KCNN4 activity in those wild-type cells. However, conferral of wild-type cation contents on HX red cells failed to restore wild-type-stimulated KCNN4 activity to those HX cells. The phenotype of reduced, maximally stimulated KCNN4 activity was shared by HX erythrocytes expressing heterozygous PIEZO1 mutants R2488Q and V598M, but not by HX erythrocytes expressing heterozygous KCNN4 mutant R352H or PIEZO1 mutant R2456H. Our data suggest that chronic KCNN4-driven red cell dehydration and intracellular cation imbalance can lead to reduced KCNN4 activity in HX and wild-type red cells.
Collapse
Affiliation(s)
- Alicia Rivera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - David H Vandorpe
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Boris E Shmukler
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnologies, "Federico II" University of Naples, CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnologies, "Federico II" University of Naples, CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Natasha M Archer
- Division of Hematology and Oncology, Boston Children's Hospital, Dana-Farber Cancer Center, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Estela Shabani
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | | | - Nelson Hamerschlak
- Department of Hematology, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - James Morton
- Quest Diagnostics, San Juan Capistrano, California
| | | | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - L Michael Snyder
- Quest Diagnostics, Marlborough, Massachusetts.,Departments of Medicine and Laboratory Medicine, University of Massachusetts Medical Center, Worcester, Massachusetts
| | - Seth L Alper
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
44
|
Harada Y, Suzuki T, Fukushige T, Kizuka Y, Yagi H, Yamamoto M, Kondo K, Inoue H, Kato K, Taniguchi N, Kanekura T, Dohmae N, Maruyama I. Generation of the heterogeneity of extracellular vesicles by membrane organization and sorting machineries. Biochim Biophys Acta Gen Subj 2019; 1863:681-691. [DOI: 10.1016/j.bbagen.2019.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/21/2018] [Accepted: 01/22/2019] [Indexed: 12/12/2022]
|
45
|
Evidence for three populations of the glucose transporter in the human erythrocyte membrane. Blood Cells Mol Dis 2019; 77:61-66. [PMID: 30974390 DOI: 10.1016/j.bcmd.2019.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/22/2019] [Accepted: 03/23/2019] [Indexed: 11/23/2022]
Abstract
Glucose transporter 1 (GLUT1) is one of 13 members of the human equilibrative glucose transport protein family and the only glucose transporter thought to be expressed in human erythrocyte membranes. Although GLUT1 has been shown to be anchored to adducin at the junctional spectrin-actin complex of the membrane through interactions with multiple proteins, whether other populations of GLUT1 also exist in the human erythrocyte membrane has not been examined. Because GLUT1 plays such a critical role in erythrocyte biology and since it comprises 10% of the total membrane protein, we undertook to evaluate the subpopulations of erythrocyte GLUT1 using single particle tracking. By monitoring the diffusion of individual AlexaFluor 488-labeled GLUT1 molecules on the surfaces of intact erythrocytes, we are able to identify three distinct subpopulations of GLUT1. While the mobility of the major subpopulation is similar to that of the anion transporter, band 3, both a more mobile and more anchored subpopulation also exist. From these studies, we conclude that ~65% of GLUT1 resides in similar or perhaps the same protein complex as band 3, while the remaining 1/3rd are either freely diffusing or interacting with other cytoskeletally anchored membrane protein complexes.
Collapse
|
46
|
Akkaya B, Kucukal E, Little JA, Gurkan UA. Mercury leads to abnormal red blood cell adhesion to laminin mediated by membrane sulfatides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1162-1171. [PMID: 30890469 DOI: 10.1016/j.bbamem.2019.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 12/27/2022]
Abstract
Exposure to mercury is associated with numerous health problems, affecting different parts of the human body, including the nervous and cardiovascular systems in adults and children; however, the underlying mechanisms are yet to be fully elucidated. We investigated the role of membrane sulfatide on mercuric ion (Hg2+) mediated red blood cell (RBC) adhesion to a sub-endothelial matrix protein, laminin, using a microfluidic system that mimics microphysiological flow conditions. We exposed whole blood to mercury (HgCl2), at a range of concentrations to mimic acute (high dose) and chronic (low dose) exposure, and examined RBC adhesion to immobilized laminin in microchannels at physiological flow conditions. Exposure of RBCs to both acute and chronic levels of Hg2+ resulted in elevated adhesive interactions between RBCs and laminin depending on the concentration of HgCl2 and exposure duration. BCAM-Lu chimer significantly inhibited the adhesion of RBCs that had been treated with 50 μM of HgCl2 solution for 1 h at 37 °C, while it did not prevent the adhesion of 3 h and 24 h Hg2+-treated RBCs. Sulfatide significantly inhibited the adhesion of RBC that had been treated with 50 μM of HgCl2 solution for 1 h at 37 °C and 0.5 μM of HgCl2 solution for 24 h at room temperature (RT). We demonstrated that RBC BCAM-Lu and RBC sulfatides bind to immobilized laminin, following exposure of RBCs to mercuric ions. The results of this study are significant considering the potential associations between sulfatides, red blood cells, mercury exposure, and cardiovascular diseases.
Collapse
Affiliation(s)
- Birnur Akkaya
- Science Faculty, Department of Molecular Biology and Genetics Department, Cumhuriyet University, Sivas, Turkey; Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Erdem Kucukal
- Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA
| | - Jane A Little
- Department of Hematology and Oncology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Umut A Gurkan
- Case Biomanufacturing and Microfabrication Laboratory, Mechanical and Aerospace Engineering Department, Case Western Reserve University, Cleveland, OH, USA; Biomedical Engineering Department, Case Western Reserve University, Cleveland, OH, USA; Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
47
|
Endocytic Markers Associated with the Internalization and Processing of Aspergillus fumigatus Conidia by BEAS-2B Cells. mSphere 2019; 4:4/1/e00663-18. [PMID: 30728282 PMCID: PMC6365614 DOI: 10.1128/msphere.00663-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Conidia from the fungus Aspergillus fumigatus are notorious for their ability to stay airborne. This characteristic is believed to allow conidia to penetrate into the cleanest environments. Several hundred conidia are thought to be inhaled each day by a given individual and then expelled by mucociliary clearance. Given that airway epithelial cells make up a significant portion of the pulmonary-air interface, we set out to determine the percentage of conidia that are actually internalized after initial contact with airway epithelial cells. We determined this through an in vitro assay using an immortalized bronchial airway epithelial cell line known as BEAS-2B. Our results suggest a small fraction of conidia are internalized by BEAS-2B cells, while the majority stay adherent to the surface of cells or are washed away during sample processing. Internalization of conidia was observed at 6 h postchallenge and not prior. Our data also indicate conidia are rendered metabolically inactive within 3 h of challenge, suggesting BEAS-2B cells process a large number of conidia without internalization in this early time frame. We have also identified several host endocytosis markers that localize around internalized conidia as well as contribute to the processing of conidia. Understanding how these host endocytosis markers affect the processing of internal and/or external conidia may provide a novel avenue for therapeutic development. Aspergillus fumigatus is a ubiquitous mold that produces small airborne conidia capable of traversing deep into the respiratory system. Recognition, processing, and clearance of A. fumigatus conidia by bronchial airway epithelial cells are thought to be relevant to host defense and immune signaling. Using z-stack confocal microscopy, we observed that only 10 to 20% of adherent conidia from the AF293 clinical isolate are internalized by BEAS-2B cells 6 h postchallenge and not prior. Similar percentages of internalization were observed for the CEA10 clinical isolate. A large subset of both AF293 and CEA10 conidia are rendered metabolically inactive without internalization at 3 h postchallenge by BEAS-2B cells. A significantly larger percentage of CEA10 conidia are metabolically active at 9 and 12 h postchallenge in comparison to the AF293 isolate, demonstrating heterogeneity among clinical isolates. We identified 7 host markers (caveolin, flotillin-2, RAB5C, RAB8B, RAB7A, 2xFYVE, and FAPP1) that consistently localized around internalized conidia 9 h postchallenge. Transient gene silencing of RAB5C, PIK3C3, and flotillin-2 resulted in a larger population of metabolically active conidia. Our findings emphasize the abundance of both host phosphatidylinositol 3-phosphate (PI3P) and PI4P around internalized conidia, as well as the importance of class III PI3P kinase for conidial processing. Therapeutic development focused on RAB5C-, PIK3C3-, and flotillin-2-mediated pathways may provide novel opportunities to modulate conidial processing and internalization. Determination of how contacted, external conidia are processed by airway epithelial cells may also provide a novel avenue to generate host-targeted therapeutics. IMPORTANCE Conidia from the fungus Aspergillus fumigatus are notorious for their ability to stay airborne. This characteristic is believed to allow conidia to penetrate into the cleanest environments. Several hundred conidia are thought to be inhaled each day by a given individual and then expelled by mucociliary clearance. Given that airway epithelial cells make up a significant portion of the pulmonary-air interface, we set out to determine the percentage of conidia that are actually internalized after initial contact with airway epithelial cells. We determined this through an in vitro assay using an immortalized bronchial airway epithelial cell line known as BEAS-2B. Our results suggest a small fraction of conidia are internalized by BEAS-2B cells, while the majority stay adherent to the surface of cells or are washed away during sample processing. Internalization of conidia was observed at 6 h postchallenge and not prior. Our data also indicate conidia are rendered metabolically inactive within 3 h of challenge, suggesting BEAS-2B cells process a large number of conidia without internalization in this early time frame. We have also identified several host endocytosis markers that localize around internalized conidia as well as contribute to the processing of conidia. Understanding how these host endocytosis markers affect the processing of internal and/or external conidia may provide a novel avenue for therapeutic development.
Collapse
|
48
|
Izquierdo I, Barrachina MN, Hermida-Nogueira L, Casas V, Eble JA, Carrascal M, Abián J, García Á. Platelet membrane lipid rafts protein composition varies following GPVI and CLEC-2 receptors activation. J Proteomics 2019; 195:88-97. [PMID: 30677554 DOI: 10.1016/j.jprot.2019.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/03/2019] [Accepted: 01/20/2019] [Indexed: 12/11/2022]
Abstract
Lipid rafts are membrane microdomains that have been proposed to play an important role in several platelet-signalling cascades, including those mediated by the receptors Glycoprotein VI (GPVI), and C-type lectin domain family 1 member B (CLEC-2), both involved in thrombus formation. We have performed a LC-MS/MS proteomic analysis of lipid rafts isolated from platelets activated through GPVI and CLEC-2 as well as from resting platelets. Our aim was to determine the magnitude of changes in lipid rafts protein composition and to elucidate the relevance of these alterations in platelet function. A number of relevant signalling proteins were found enriched in lipid rafts following platelet activation (such as the tyrosine protein kinases Fyn, Lyn and Yes; the G proteins G(i) and G(z); and cAMP protein kinase). Interestingly, our results indicate that the relative enrichment of lipid rafts in these signalling proteins may not be a consequence of protein translocation to these domains upon platelet stimulation, but the result of a massive loss in cytoskeletal proteins after platelet activation. Thus, this study may help to better understand the effects of platelet activation in the reorganization of lipid rafts and set the basis for further proteomic studies of these membrane microdomains in platelets. SIGNIFICANCE: We performed the first proteomic comparative analysis of lipid rafts- protein composition in platelets activated through GPVI and CLEC-2 receptors and in resting state. We identified a number of signalling proteins essential for platelet activation relatively enriched in platelets activated through both receptors, and we show that lipid rafts reorganization upon platelet activation leads to a loss in cytoskeletal proteins, highly associated to these domains in resting platelets.
Collapse
Affiliation(s)
- Irene Izquierdo
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - María N Barrachina
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Lidia Hermida-Nogueira
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Vanessa Casas
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC-IDIBAPS, Barcelona, Spain
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | | | - Joaquín Abián
- CSIC/UAB Proteomics Laboratory, IIBB-CSIC-IDIBAPS, Barcelona, Spain
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
49
|
Ghafarian F, Pashirzad M, Khazaei M, Rezayi M, Hassanian SM, Ferns GA, Avan A. The clinical impact of exosomes in cardiovascular disorders: From basic science to clinical application. J Cell Physiol 2018; 234:12226-12236. [PMID: 30536994 DOI: 10.1002/jcp.27964] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease (CVD) is the major cause of death globally; therefore, there is a need for the identification of a valid biomarker that accurately predicts the risk of developing CVD, and novel therapeutic approaches for its treatment. Exosomes are very small extracellular vesicles containing protein, lipid, transcription factors, messenger RNAs, noncoding RNA, and nucleic acid contents that are important players in intercellular communication, and that act via long-range signals or cell-to-cell contact. The discovery of exosomes provides potential strategies for the diagnosis and treatment of CVD. In the current review, we have explored the potential impact of exosomes on cardiovascular physiology, and their therapeutic potential in cardiovascular disorders with an emphasis on the existing preclinical studies.
Collapse
Affiliation(s)
- Farzaneh Ghafarian
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Pashirzad
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezayi
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Amir Avan
- Department of Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
50
|
McNamara RP, Caro-Vegas CP, Costantini LM, Landis JT, Griffith JD, Damania BA, Dittmer DP. Large-scale, cross-flow based isolation of highly pure and endocytosis-competent extracellular vesicles. J Extracell Vesicles 2018; 7:1541396. [PMID: 30533204 PMCID: PMC6282418 DOI: 10.1080/20013078.2018.1541396] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/02/2018] [Accepted: 10/12/2018] [Indexed: 12/15/2022] Open
Abstract
Isolation of extracellular vesicles (EVs) from cell culture supernatant or plasma can be accomplished in a variety of ways. Common measures to quantify relative success are: concentration of the EVs, purity from non-EVs associated protein, size homogeneity and functionality of the final product. Here, we present an industrial-scale workflow for isolating highly pure and functional EVs using cross-flow based filtration coupled with high-molecular weight Capto Core size exclusion. Through this combination, EVs loss is kept to a minimum. It outperforms other isolation procedures based on a number of biochemical and biophysical assays. Moreover, EVs isolated through this method can be further concentrated down or directly immunopurified to obtain discreet populations of EVs. From our results, we propose that cross-flow/Capto Core isolation is a robust method of purifying highly concentrated, homogenous, and functionally active EVs from industrial-scale input volumes with few contaminants relative to other methods.
Collapse
Affiliation(s)
- Ryan P. McNamara
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Carolina P. Caro-Vegas
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Lindsey M. Costantini
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Justin T. Landis
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Jack D. Griffith
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Blossom A. Damania
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|