1
|
Pagliuca C, Colicchio R, Resta SC, Talà A, Scaglione E, Mantova G, Continisio L, Pagliarulo C, Bucci C, Alifano P, Salvatore P. Neisseria meningitidis activates pyroptotic pathways in a mouse model of meningitis: role of a two-partner secretion system. Front Cell Infect Microbiol 2024; 14:1384072. [PMID: 39376663 PMCID: PMC11456522 DOI: 10.3389/fcimb.2024.1384072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024] Open
Abstract
There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1β and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.
Collapse
Affiliation(s)
- Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Mantova
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Leonardo Continisio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Caterina Pagliarulo
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Pietro Alifano
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- The Institute CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
- Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
2
|
Rasl J, Caslavsky J, Grusanovic J, Chvalova V, Kosla J, Adamec J, Grousl T, Klimova Z, Vomastek T. Depletion of calpain2 accelerates epithelial barrier establishment and reduces growth factor-induced cell scattering. Cell Signal 2024; 121:111295. [PMID: 38996955 DOI: 10.1016/j.cellsig.2024.111295] [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: 10/24/2023] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Calpain2 is a conventional member of the non-lysosomal calpain protease family that has been shown to affect the dynamics of focal and cell-cell adhesions by proteolyzing the components of adhesion complexes. Here, we inactivated calpain2 using CRISPR/Cas9 in epithelial MDCK cells. We show that depletion of calpain2 has multiple effects on cell morphology and function. Calpain2-depleted cells develop epithelial shape, however, they cover a smaller area, and cell clusters are more compact. Inactivation of calpain2 enhanced restoration of transepithelial electrical resistance after calcium switch, decreased cell migration, and delayed cell scattering induced by HGF/SF. In addition, calpain2 depletion prevented morphological changes induced by ERK2 overexpression. Interestingly, proteolysis of several calpain2 targets, including E-cadherin, β-catenin, talin, FAK, and paxillin, was not discernibly affected by calpain2 depletion. Taken together, these data suggest that calpain2 regulates the stability of cell-cell and cell-substratum adhesions indirectly without affecting the proteolysis of these adhesion complexes.
Collapse
Affiliation(s)
- Jan Rasl
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Josef Caslavsky
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Josipa Grusanovic
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vera Chvalova
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jan Kosla
- Laboratory of Viral and Cellular Genetics and Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Jiri Adamec
- Department of Interdisciplinary Oncology, Louisiana State University HSC School of Medicine, New Orleans, USA
| | - Tomas Grousl
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Zuzana Klimova
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic.
| | - Tomas Vomastek
- Laboratory of Cell Signalling Institute of Microbiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic.
| |
Collapse
|
3
|
Cheng D, Wang J, Yao M, Cox CD. Joining forces: crosstalk between mechanosensitive PIEZO1 ion channels and integrin-mediated focal adhesions. Biochem Soc Trans 2023; 51:1897-1906. [PMID: 37772664 DOI: 10.1042/bst20230042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023]
Abstract
Both integrin-mediated focal adhesions (FAs) and mechanosensitive ion channels such as PIEZO1 are critical in mechanotransduction processes that influence cell differentiation, development, and cancer. Ample evidence now exists for regulatory crosstalk between FAs and PIEZO1 channels with the molecular mechanisms underlying this process remaining unclear. However, an emerging picture is developing based on spatial crosstalk between FAs and PIEZO1 revealing a synergistic model involving the cytoskeleton, extracellular matrix (ECM) and calcium-dependent signaling. Already cell type, cell contractility, integrin subtypes and ECM composition have been shown to regulate this crosstalk, implying a highly fine-tuned relationship between these two major mechanosensing systems. In this review, we summarize the latest advances in this area, highlight the physiological implications of this crosstalk and identify gaps in our knowledge that will improve our understanding of cellular mechanosensing.
Collapse
Affiliation(s)
- Delfine Cheng
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Junfan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
| | - Charles D Cox
- The Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Kensington, NSW 2052, Australia
| |
Collapse
|
4
|
Suzuki M, Takeshita K, Kitamura Y, Kuribayashi M, Huang Z, Ichihara G, Oikawa S, Ichihara S. In Vitro Exposure to Glucose Alters the Expression of Phosphorylated Proteins in Platelets. Biomedicines 2023; 11:biomedicines11020543. [PMID: 36831080 PMCID: PMC9953272 DOI: 10.3390/biomedicines11020543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Diabetes mellitus (DM) is a pro-thrombotic state that can potentially cause serious cardiovascular complications. Platelet hyperactivation plays an important role in these pathological processes, however there is little or no information on the effect of hyperglycemia on platelet proteins. The aim of this study was to identify the molecular targets associated with platelet reactivity under hyperglycemia. Towards this goal, we examined the effects of the exposure of platelets to 1 and 2 h glucose (300 mg/dL) and control (vehicle and osmolality control using mannitol) on platelet proteins (n = 4 samples per group) using two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) combined with MALDI-TOF/TOF tandem mass spectrometry. Two-hour exposure to glucose significantly up-regulated the expression of ATP synthase subunit beta, filamin-A, and L-lactate dehydrogenase A chain in platelets. Pro-Q Diamond staining confirmed the effect of 2 h glucose on vinculin, heat shock protein HSP 90-alpha, filamin-A, and fructose-bisphosphate aldolase A (platelet phosphorylated proteins). The identified proteins are involved in various cellular processes and functions and possibly in platelet reactivity under hyperglycemic conditions.
Collapse
Affiliation(s)
- Mizuho Suzuki
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
| | - Kyosuke Takeshita
- Department of Clinical Laboratory, Saitama Medical Center, Saitama University, Saitama 350-8550, Japan
| | - Yuki Kitamura
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
| | - Marie Kuribayashi
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu 514-8507, Japan
| | - Zhenlie Huang
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu 514-8507, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke 329-0498, Japan
- Department of Human Functional Genomics, Life Science Research Center, Mie University, Tsu 514-8507, Japan
- Correspondence:
| |
Collapse
|
5
|
Shih YT, Wei SY, Chen JH, Wang WL, Wu HY, Wang MC, Lin CY, Lee PL, Lin CY, Chiang HC, Chen YJ, Chien S, Chiu JJ. Vinculin phosphorylation impairs vascular endothelial junctions promoting atherosclerosis. Eur Heart J 2023; 44:304-318. [PMID: 36380599 PMCID: PMC10202442 DOI: 10.1093/eurheartj/ehac647] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 08/26/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND AIMS Atherosclerosis preferentially develops in arterial branches and curvatures where vascular endothelium is exposed to disturbed flow. In this study, the effects of disturbed flow on the regulation of vascular endothelial phosphoproteins and their contribution to therapeutic application in atherogenesis were elucidated. METHODS Porcine models, large-scale phosphoproteomics, transgenic mice, and clinical specimens were used to discover novel site-specific phosphorylation alterations induced by disturbed flow in endothelial cells (ECs). RESULTS A large-scale phosphoproteomics analysis of native endothelium from disturbed (athero-susceptible) vs. pulsatile flow (athero-resistant) regions of porcine aortas led to the identification of a novel atherosclerosis-related phosphoprotein vinculin (VCL) with disturbed flow-induced phosphorylation at serine 721 (VCLS721p). The induction of VCLS721p was mediated by G-protein-coupled receptor kinase 2 (GRK2)S29p and resulted in an inactive form of VCL with a closed conformation, leading to the VE-cadherin/catenin complex disruption to enhance endothelial permeability and atherogenesis. The generation of novel apolipoprotein E-deficient (ApoE-/-) mice overexpressing S721-non-phosphorylatable VCL mutant in ECs confirmed the critical role of VCLS721p in promoting atherosclerosis. The administration of a GRK2 inhibitor to ApoE-/- mice suppressed plaque formation by inhibiting endothelial VCLS721p. Studies on clinical specimens from patients with coronary artery disease (CAD) revealed that endothelial VCLS721p is a critical clinicopathological biomarker for atherosclerosis progression and that serum VCLS721p level is a promising biomarker for CAD diagnosis. CONCLUSIONS The findings of this study indicate that endothelial VCLS721p is a valuable hemodynamic-based target for clinical assessment and treatment of vascular disorders resulting from atherosclerosis.
Collapse
Affiliation(s)
- Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Shu-Yi Wei
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jin-Hua Chen
- Graduate Institute of Data Science, College of Management, Health Data Analytics and Statistics Center, Office of Data Science, Biostatistics Center, Department of Medical Research, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Li Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, Taipei 10617, Taiwan
| | - Mei-Cun Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chia-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Pei-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Hung-Che Chiang
- Department of Pharmacy, School of Pharmacy, China Medical University, Taichung 404327, Taiwan
| | - Yu-Ju Chen
- Academic Sinica, Institute of Chemistry, Taipei 11529, Taiwan
| | - Shu Chien
- Departments of Bioengineering and Medicine, and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
- School of Medical Laboratory Science and Biotechnology, Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30071, Taiwan
| |
Collapse
|
6
|
Vibration, a treatment for migraine, linked to calpain driven changes in actin cytoskeleton. PLoS One 2022; 17:e0262058. [PMID: 35482731 PMCID: PMC9049534 DOI: 10.1371/journal.pone.0262058] [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: 12/14/2021] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding how a human cell reacts to external physical stimuli is essential to understanding why vibration can elicit localized pain reduction. Stimulation of epithelial cells with external vibration forces has been shown to change cell shape, particularly in regards to structures involved in non-muscle cell motility. We hypothesized that epithelial cells respond to vibration transduction by altering proteins involved in remodeling cytoskeleton. Epithelial cells were exposed to vibration and assessed by microscopy, cytoskeletal staining, immunoblotting and quantitative RT-PCR. Here, we report that epithelial cell lines exposed to 15 minutes of vibration retract filopodia and concentrate actin at the periphery of the cell. In particular, we show an increased expression of the calcium-dependent, cysteine protease, calpain. The discovery that cell transitions are induced by limited exposure to natural forces, such as vibration, provides a foundation to explain how vibrational treatment helps migraine patients.
Collapse
|
7
|
Abstract
INTRODUCTION Calpain-1 and calpain-2 are prototypical classical isoforms of the calpain family of calcium-activated cysteine proteases. Their substrate proteins participate in a wide range of cellular processes, including transcription, survival, proliferation, apoptosis, migration, and invasion. Dysregulated calpain activity has been implicated in tumorigenesis, suggesting that calpains may be promising therapeutic targets. AREAS COVERED This review covers clinical and basic research studies implicating calpain-1 and calpain-2 expression and activity in tumorigenesis and metastasis. We highlight isoform specific functions and provide an overview of substrates and cancer-related signalling pathways affected by calpain-mediated proteolytic cleavage. We also discuss efforts to develop clinically relevant calpain specific inhibitors and spotlight the challenges facing inhibitor development. EXPERT OPINION Rationale for targeting calpain-1 and calpain-2 in cancer is supported by pre-clinical and clinical studies demonstrating that calpain inhibition has the potential to attenuate carcinogenesis and block metastasis of aggressive tumors. The wide range of substrates and cleavage products, paired with inconsistencies in model systems, underscores the need for more complete understanding of physiological substrates and how calpain cleavage alters their function in cellular processes. The development of isoform specific calpain inhibitors remains an important goal with therapeutic potential in cancer and other diseases.
Collapse
Affiliation(s)
- Ivan Shapovalov
- Department of Pathology and Molecular Medicine, Queen's University, Division of Cancer Biology and Genetics, Queen's Cancer Research Institute, 10 Stuart Street, Botterell Hall, Room A309, Kingston, Ontario, K7L 3N6 Canada
| | - Danielle Harper
- Department of Pathology and Molecular Medicine, Queen's University, Division of Cancer Biology and Genetics, Queen's Cancer Research Institute, 10 Stuart Street, Botterell Hall, Room A309, Kingston, Ontario, K7L 3N6 Canada
| | - Peter A Greer
- Department of Pathology and Molecular Medicine, Queen's University, Division of Cancer Biology and Genetics, Queen's Cancer Research Institute, 10 Stuart Street, Botterell Hall, Room A309, Kingston, Ontario, K7L 3N6 Canada
| |
Collapse
|
8
|
Lin YS, Lin YH, Nguyen Thi M, Hsiao SC, Chiu WT. STIM1 Controls the Focal Adhesion Dynamics and Cell Migration by Regulating SOCE in Osteosarcoma. Int J Mol Sci 2021; 23:ijms23010162. [PMID: 35008585 PMCID: PMC8745645 DOI: 10.3390/ijms23010162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/12/2022] Open
Abstract
The dysregulation of store-operated Ca2+ entry (SOCE) promotes cancer progression by changing Ca2+ levels in the cytosol or endoplasmic reticulum. Stromal interaction molecule 1 (STIM1), a component of SOCE, is upregulated in several types of cancer and responsible for cancer cell migration, invasion, and metastasis. To explore the impact of STIM1-mediated SOCE on the turnover of focal adhesion (FA) and cell migration, we overexpressed the wild-type and constitutively active or dominant negative variants of STIM1 in an osteosarcoma cell line. In this study, we hypothesized that STIM1-mediated Ca2+ elevation may increase cell migration. We found that constitutively active STIM1 dramatically increased the Ca2+ influx, calpain activity, and turnover of FA proteins, such as the focal adhesion kinase (FAK), paxillin, and vinculin, which impede the cell migration ability. In contrast, dominant negative STIM1 decreased the turnover of FA proteins as its wild-type variant compared to the cells without STIM1 overexpression while promoting cell migration. These unexpected results suggest that cancer cells need an appropriate amount of Ca2+ to control the assembly and disassembly of focal adhesions by regulating calpain activity. On the other hand, overloaded Ca2+ results in excessive calpain activity, which is not beneficial for cancer metastasis.
Collapse
Affiliation(s)
- Yu-Shan Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan;
| | - Yi-Hsin Lin
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; (Y.-H.L.); (M.N.T.)
| | - MyHang Nguyen Thi
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; (Y.-H.L.); (M.N.T.)
| | - Shih-Chuan Hsiao
- Department of Hematology & Oncology, Saint Martin de Porres Hospital, Chiayi 600, Taiwan;
| | - Wen-Tai Chiu
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan;
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; (Y.-H.L.); (M.N.T.)
- Medical Device Innovation Center, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence:
| |
Collapse
|
9
|
Liu L, Zhang Q, Xiao S, Sun Z, Ding S, Chen Y, Wang L, Yin X, Liao F, Jiang LH, Xue M, You Y. Inhibition of Shear-Induced Platelet Aggregation by Xueshuantong via Targeting Piezo1 Channel-Mediated Ca 2+ Signaling Pathway. Front Pharmacol 2021; 12:606245. [PMID: 33841141 PMCID: PMC8025832 DOI: 10.3389/fphar.2021.606245] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
XueShuanTong (XST) comprising therapeutically active ginsenosides, a lyophilized extract of Panax notoginseng roots, is extensively used in traditional Chinese medicine to treat ischemic heart and cerebrovascular diseases. Our recent study shows that treatment with XST inhibits shear-induced thrombosis formation but the underlying mechanism remained unclear. This study aimed to investigate the hypothesis that XST inhibited shear-induced platelet aggregation via targeting the mechanosensitive Ca2+-permeable Piezo1 channel by performing platelet aggregation assay, Ca2+ imaging and Western blotting analysis. Exposure to shear at physiologically (1,000–2000 s−1) and pathologically related rates (4,000–6,000 s−1) induced platelet aggregation that was inhibited by treatment with GsMTx-4. Exposure to shear evoked robust Ca2+ responses in platelets that were inhibited by treatment with GsMTx-4 and conversely enhanced by treatment with Yoda1. Treatment with XST at a clinically relevant concentration (0.15 g L−1) potently inhibited shear-induced Ca2+ responses and platelet aggregation, without altering vWF-mediated platelet adhesion and rolling. Exposure to shear, while resulting in no effect on the calpain-2 expression in platelets, induced calpain-2-mediated cleavage of talin1 protein, which is known to be critical for platelet activation. Shear-induced activation of calpain-2 and cleavage of talin1 were attenuated by treatment with XST. Taken together, our results suggest that XST inhibits shear-induced platelet aggregation via targeting the Piezo1 channel to prevent Piezo1-mediated Ca2+ signaling and downstream calpain-2 and talin1 signal pathway, thus providing novel insights into the mechanism of the therapeutic action of XST on platelet aggregation and thrombosis formation.
Collapse
Affiliation(s)
- Lei Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiongling Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shunli Xiao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengxiao Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shilan Ding
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lan Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojie Yin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fulong Liao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury, Xinxiang Medical University, Xinxiang, China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Mei Xue
- XiYuan Hosipital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yun You
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Manipulation of Focal Adhesion Signaling by Pathogenic Microbes. Int J Mol Sci 2021; 22:ijms22031358. [PMID: 33572997 PMCID: PMC7866387 DOI: 10.3390/ijms22031358] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Abstract
Focal adhesions (FAs) serve as dynamic signaling hubs within the cell. They connect intracellular actin to the extracellular matrix (ECM) and respond to environmental cues. In doing so, these structures facilitate important processes such as cell-ECM adhesion and migration. Pathogenic microbes often modify the host cell actin cytoskeleton in their pursuit of an ideal replicative niche or during invasion to facilitate uptake. As actin-interfacing structures, FA dynamics are also intimately tied to actin cytoskeletal organization. Indeed, exploitation of FAs is another avenue by which pathogenic microbes ensure their uptake, survival and dissemination. This is often achieved through the secretion of effector proteins which target specific protein components within the FA. Molecular mimicry of the leucine-aspartic acid (LD) motif or vinculin-binding domains (VBDs) commonly found within FA proteins is a common microbial strategy. Other effectors may induce post-translational modifications to FA proteins through the regulation of phosphorylation sites or proteolytic cleavage. In this review, we present an overview of the regulatory mechanisms governing host cell FAs, and provide examples of how pathogenic microbes have evolved to co-opt them to their own advantage. Recent technological advances pose exciting opportunities for delving deeper into the mechanistic details by which pathogenic microbes modify FAs.
Collapse
|
11
|
Tian X, Inoue K, Zhang Y, Wang Y, Sperati CJ, Pedigo CE, Zhao T, Yan M, Groener M, Moledina DG, Ebenezer K, Li W, Zhang Z, Liebermann DA, Greene L, Greer P, Parikh CR, Ishibe S. Inhibiting calpain 1 and 2 in cyclin G associated kinase-knockout mice mitigates podocyte injury. JCI Insight 2020; 5:142740. [PMID: 33208557 PMCID: PMC7710277 DOI: 10.1172/jci.insight.142740] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/08/2020] [Indexed: 12/27/2022] Open
Abstract
Evidence for reduced expression of cyclin G associated kinase (GAK) in glomeruli of patients with chronic kidney disease was observed in the Nephroseq human database, and GAK was found to be associated with the decline in kidney function. To examine the role of GAK, a protein that functions to uncoat clathrin during endocytosis, we generated podocyte-specific Gak-knockout mice (Gak-KO), which developed progressive proteinuria and kidney failure with global glomerulosclerosis. We isolated glomeruli from the mice carrying the mutation to perform messenger RNA profiling and unearthed evidence for dysregulated podocyte calpain protease activity as an important contributor to progressive podocyte damage. Treatment with calpain inhibitor III specifically inhibited calpain-1/-2 activities, mitigated the degree of proteinuria and glomerulosclerosis, and led to a striking increase in survival in the Gak-KO mice. Podocyte-specific deletion of Capns1, essential for calpain-1 and calpain-2 activities, also improved proteinuria and glomerulosclerosis in Gak-KO mice. Increased podocyte calpain activity-mediated proteolysis of IκBα resulted in increased NF-κB p65-induced expression of growth arrest and DNA-damage-inducible 45 beta in the Gak-KO mice. Our results suggest that loss of podocyte-associated Gak induces glomerular injury secondary to calcium dysregulation and aberrant calpain activation, which when inhibited, can provide a protective role.
Collapse
MESH Headings
- Animals
- Calpain/antagonists & inhibitors
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/therapy
- Female
- Glomerulosclerosis, Focal Segmental/etiology
- Glomerulosclerosis, Focal Segmental/metabolism
- Glomerulosclerosis, Focal Segmental/pathology
- Glomerulosclerosis, Focal Segmental/therapy
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Podocytes/metabolism
- Podocytes/pathology
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/physiology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/therapy
Collapse
Affiliation(s)
- Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yan Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - C. John Sperati
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher E. Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tingting Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Meihua Yan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marwin Groener
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dennis G. Moledina
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Karen Ebenezer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Wei Li
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhenhai Zhang
- State Key Laboratory of Organ Failure Research, Southern Medical University, Nanfang Hospital, Guangzhou, China
- Center for Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dan A. Liebermann
- Fels Institute of Cancer Research and Molecular Biology and Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania USA
| | - Lois Greene
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Peter Greer
- Queen’s Cancer Research Institute, Kingston, Ontario, Canada
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Miyazaki T, Miyazaki A. Dysregulation of Calpain Proteolytic Systems Underlies Degenerative Vascular Disorders. J Atheroscler Thromb 2017; 25:1-15. [PMID: 28819082 PMCID: PMC5770219 DOI: 10.5551/jat.rv17008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic vascular diseases such as atherosclerosis, aneurysms, diabetic angiopathy/retinopathy as well as fibrotic and proliferative vascular diseases are generally complicated by the progression of degenerative insults, which are characterized by endothelial dysfunction, apoptotic/necrotic cell death in vascular/immune cells, remodeling of extracellular matrix or breakdown of elastic lamella. Increasing evidence suggests that dysfunctional calpain proteolytic systems and defective calpain protein metabolism in blood vessels contribute to degenerative disorders. In vascular endothelial cells, the overactivation of conventional calpains consisting of calpain-1 and -2 isozymes can lead to the disorganization of cell-cell junctions, dysfunction of nitric oxide synthase, sensitization of Janus kinase/signal transducer and activator of transcription cascades and depletion of prostaglandin I2, which contributes to degenerative disorders. In addition to endothelial cell dysfunctions, calpain overactivation results in inflammatory insults in macrophages and excessive fibrogenic/proliferative signaling in vascular smooth muscle cells. Moreover, calpain-6, a non-proteolytic unconventional calpain, is involved in the conversion of macrophages to a pro-atherogenic phenotype, leading to the pinocytotic deposition of low-density lipoprotein cholesterol in the cells. Here, we discuss the recent progress that has been made in our understanding of how calpain contributes to degenerative vascular disorders.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine
| |
Collapse
|
13
|
Miyazaki T, Miyazaki A. Emerging roles of calpain proteolytic systems in macrophage cholesterol handling. Cell Mol Life Sci 2017; 74:3011-3021. [PMID: 28432377 PMCID: PMC11107777 DOI: 10.1007/s00018-017-2528-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 01/21/2023]
Abstract
Calpains are Ca2+-dependent intracellular proteases that play central roles in the post-translational processing of functional proteins. In mammals, calpain proteolytic systems comprise the endogenous inhibitor calpastatin as well as 15 homologues of the catalytic subunits and two homologues of the regulatory subunits. Recent pharmacological and gene targeting studies in experimental animal models have revealed the contribution of conventional calpains, which consist of the calpain-1 and -2 isozymes, to atherosclerotic diseases. During atherogenesis, conventional calpains facilitate the CD36-dependent uptake of oxidized low-density lipoprotein (LDL), and block cholesterol efflux through ATP-binding cassette transporters in lesional macrophages, allowing the expansion of lipid-enriched atherosclerotic plaques. In addition, calpain-6, an unconventional non-proteolytic calpain, in macrophages reportedly potentiates pinocytotic uptake of native LDL, and attenuates the efferocytic clearance of apoptotic and necrotic cell corpses from the lesions. Herein, we discuss the recent progress that has been made in our understanding of how calpain contributes to atherosclerosis, in particular focusing on macrophage cholesterol handling.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| |
Collapse
|
14
|
Nourse JL, Pathak MM. How cells channel their stress: Interplay between Piezo1 and the cytoskeleton. Semin Cell Dev Biol 2017; 71:3-12. [PMID: 28676421 DOI: 10.1016/j.semcdb.2017.06.018] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 11/17/2022]
Abstract
Cells constantly encounter mechanical stimuli in their environment, such as dynamic forces and mechanical features of the extracellular matrix. These mechanical cues are transduced into biochemical signals, and integrated with genetic and chemical signals to modulate diverse physiological processes. Cells also actively generate forces to internally transport cargo, to explore the physical properties of their environment and to spatially position themselves and other cells during development. Mechanical forces are therefore central to development, homeostasis, and repair. Several molecular and biophysical strategies are utilized by cells for detecting and generating mechanical forces. Here we discuss an important class of molecules involved in sensing and transducing mechanical forces - mechanically-activated ion channels. We focus primarily on the Piezo1 ion channel, and examine its relationship with the cellular cytoskeleton.
Collapse
Affiliation(s)
- Jamison L Nourse
- Department of Physiology & Biophysics, Sue & Bill Gross Stem Cell Research Center, 835 Health Sciences Road, Room 275B, UC Irvine, Irvine, CA 92697, United States
| | - Medha M Pathak
- Department of Physiology & Biophysics, Sue & Bill Gross Stem Cell Research Center, 835 Health Sciences Road, Room 275B, UC Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
15
|
Coly PM, Gandolfo P, Castel H, Morin F. The Autophagy Machinery: A New Player in Chemotactic Cell Migration. Front Neurosci 2017; 11:78. [PMID: 28261054 PMCID: PMC5311050 DOI: 10.3389/fnins.2017.00078] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/03/2017] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved self-degradative process that plays a key role in diverse cellular processes such as stress response or differentiation. A growing body of work highlights the direct involvement of autophagy in cell migration and cancer metastasis. Specifically, autophagy has been shown to be involved in modulating cell adhesion dynamics as well as epithelial-to-mesenchymal transition. After providing a general overview of the mechanisms controlling autophagosome biogenesis and cell migration, we discuss how chemotactic G protein-coupled receptors, through the repression of autophagy, may orchestrate membrane trafficking and compartmentation of specific proteins at the cell front in order to support the critical steps of directional migration.
Collapse
Affiliation(s)
- Pierre-Michaël Coly
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale (INSERM), DC2NRouen, France; Institute for Research and Innovation in BiomedicineRouen, France
| | - Pierrick Gandolfo
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale (INSERM), DC2NRouen, France; Institute for Research and Innovation in BiomedicineRouen, France
| | - Hélène Castel
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale (INSERM), DC2NRouen, France; Institute for Research and Innovation in BiomedicineRouen, France
| | - Fabrice Morin
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale (INSERM), DC2NRouen, France; Institute for Research and Innovation in BiomedicineRouen, France
| |
Collapse
|
16
|
Chang SJ, Chen YC, Yang CH, Huang SC, Huang HK, Li CC, Harn HIC, Chiu WT. Revealing the three dimensional architecture of focal adhesion components to explain Ca 2+-mediated turnover of focal adhesions. Biochim Biophys Acta Gen Subj 2017; 1861:624-635. [PMID: 28063985 DOI: 10.1016/j.bbagen.2017.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Focal adhesions (FAs) are large, dynamic protein complexes located close to the plasma membrane, which serve as the mechanical linkages and a biochemical signaling hub of cells. The coordinated and dynamic regulation of focal adhesion is required for cell migration. Degradation, or turnover, of FAs is a major event at the trailing edge of a migratory cell, and is mediated by Ca2+/calpain-dependent proteolysis and disassembly. Here, we investigated how Ca2+ influx induces cascades of FA turnover in living cells. METHODS Images obtained with a total internal reflection fluorescence microscope (TIRFM) showed that Ca2+ ions induce different processes in the FA molecules focal adhesion kinase (FAK), paxillin, vinculin, and talin. Three mutated calpain-resistant FA molecules, FAK-V744G, paxillin-S95G, and talin-L432G, were used to clarify the role of each FA molecule in FA turnover. RESULTS Vinculin was resistant to degradation and was not significantly affected by the presence of mutated calpain-resistant FA molecules. In contrast, talin was more sensitive to calpain-mediated turnover than the other molecules. Three-dimensional (3D) fluorescence imaging and immunoblotting demonstrated that outer FA molecules were more sensitive to calpain-mediated proteolysis than internal FA molecules. Furthermore, cell contraction is not involved in degradation of FA. CONCLUSIONS These results suggest that Ca2+-mediated degradation of FAs was mediated by both proteolysis and disassembly. The 3D architecture of FAs is related to the different dynamics of FA molecule degradation during Ca2+-mediated FA turnover. GENERAL SIGNIFICANCE This study will help us to clearly understand the underlying mechanism of focal adhesion turnover by Ca2+.
Collapse
Affiliation(s)
- Shu-Jing Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Ying-Chi Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Hsun Yang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Soon-Cen Huang
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Liouying Campus, Tainan 736, Taiwan
| | - Ho-Kai Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Chun-Chun Li
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Hans I-Chen Harn
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
17
|
Thorpe H, Akhlaq M, Jackson D, Al Ghamdi S, Storr S, Martin S, Ilyas M. Multiple pathways regulate Cten in colorectal cancer without a Tensin switch. Int J Exp Pathol 2016; 96:362-9. [PMID: 26852686 DOI: 10.1111/iep.12154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023] Open
Abstract
CTEN/TNS4 is a member of the Tensin gene family. It localizes to focal adhesions and induces cell motility. The mechanisms regulating Cten expression are unclear although we have shown regulation by Kras in the colon and pancreas. In normal mammary cell lines, it is reportedly upregulated by epidermal growth factor receptor (EGFR) and STAT3 signalling and upregulation is accompanied by downregulation of Tensin 3 (Tensin switch). In this study, we investigated the roles of EGFR and STAT3 signalling in the regulation of Cten in colorectal cancer (CRC). In addition, we investigated calpain--a regulator of focal adhesion-associated proteins whose relevance to Cten has not been investigated. CRC cell lines were stimulated with epidermal growth factor (EGF). This resulted in an increase in Cten and Tensin 3 protein. Kras was knocked down and this resulted in downregulation of Cten and Tensin 3. We next investigated the role of STAT3 signalling. Activation and knockdown of STAT3 resulted in downregulation and upregulation, respectively, of Cten. Inhibition of calpain resulted in upregulation of both Cten and Tensin 3. As the regulators of Cten also seemed to regulate Tensin 3, we tested the interaction between Cten and Tensin 3. Cten was forcibly expressed or knocked down resulting, respectively, in upregulation and downregulation of Tensin 3. We conclude that in CRC, Cten is upregulated by EGFR and Kras but downregulated by STAT3. We show that calpain may be a negative regulator of Cten and that a Tensin switch does not occur and, if anything, Cten stabilizes Tensin 3.
Collapse
Affiliation(s)
- Hannah Thorpe
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Maham Akhlaq
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Darryl Jackson
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Saleh Al Ghamdi
- King Abdullah International Medical Research Center, KSAU-HS, Riyadh, Saudi Arabia
| | - Sarah Storr
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Stewart Martin
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Mohammad Ilyas
- School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
18
|
Schubert P, Coupland D, Nombalais M, M Walsh G, Devine DV. RhoA/ROCK signaling contributes to sex differences in the activation of human platelets. Thromb Res 2016; 139:50-5. [PMID: 26916296 DOI: 10.1016/j.thromres.2016.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/18/2015] [Accepted: 01/10/2016] [Indexed: 02/08/2023]
Abstract
Studies of sex-dependent differences in platelet aggregation and glycoprotein (GP)IIb/IIIa activation have demonstrated that platelets from females are more sensitive to agonists than those from males. To date, there is little understanding of these differences at a molecular level. Here, sex differences in reactivity of platelets from 86 women and 86 men were investigated. Platelet degranulation (CD62P expression) and activation of GPIIb/IIIa (PAC-1 binding), with and without ADP, were assessed. Extent of shape change (ESC) in response to ADP was measured. Basal CD62P and PAC-1 expression did not differ between the sexes. In response to ADP activation, mean PAC-1 binding in platelets from female donors was 17.9±3.5% vs. 14.0±4.1% in platelets from male donors, and ESC was significantly greater in platelets from females (p<0.05). Evaluation of basal expression of signaling molecules along the ADP receptor pathway leading to GPIIb/IIIa activation and subsequent RhoA/ROCK signaling via GPIIb/IIIa 'outside-in' signaling showed that platelets from females produce 3-fold greater levels of phosphorylated protein kinase C (PKC) substrates. There was a 2.5-fold greater level of activated RhoA, and platelet sub-fractionation analysis demonstrated 2.7-fold more RhoA in the membrane fraction of female vs. male platelets. Similarly, there was a 2.8-fold increase in levels of phosphorylated myosin light chain (MLC) in platelets from females vs. males. The increased signaling activity in platelets from females mirrors their greater sensitivity to agonists. These findings further our understanding of the molecular differences between platelets from males and females.
Collapse
Affiliation(s)
- Peter Schubert
- Centre for Innovation, Canadian Blood Services, Vancouver, BC, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Danielle Coupland
- Centre for Innovation, Canadian Blood Services, Vancouver, BC, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Marie Nombalais
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Geraldine M Walsh
- Centre for Innovation, Canadian Blood Services, Vancouver, BC, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Dana V Devine
- Centre for Innovation, Canadian Blood Services, Vancouver, BC, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
19
|
Bettaieb A, Averill-Bates DA. Thermotolerance induced at a mild temperature of 40°C alleviates heat shock-induced ER stress and apoptosis in HeLa cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:52-62. [DOI: 10.1016/j.bbamcr.2014.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 12/30/2022]
|
20
|
Moretti D, Del Bello B, Allavena G, Maellaro E. Calpains and cancer: Friends or enemies? Arch Biochem Biophys 2014; 564:26-36. [DOI: 10.1016/j.abb.2014.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 09/23/2014] [Accepted: 09/30/2014] [Indexed: 02/07/2023]
|
21
|
Tian X, Kim JJ, Monkley SM, Gotoh N, Nandez R, Soda K, Inoue K, Balkin DM, Hassan H, Son SH, Lee Y, Moeckel G, Calderwood DA, Holzman LB, Critchley DR, Zent R, Reiser J, Ishibe S. Podocyte-associated talin1 is critical for glomerular filtration barrier maintenance. J Clin Invest 2014; 124:1098-113. [PMID: 24531545 PMCID: PMC3934159 DOI: 10.1172/jci69778] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022] Open
Abstract
Podocytes are specialized actin-rich epithelial cells that line the kidney glomerular filtration barrier. The interface between the podocyte and the glomerular basement membrane requires integrins, and defects in either α3 or β1 integrin, or the α3β1 ligand laminin result in nephrotic syndrome in murine models. The large cytoskeletal protein talin1 is not only pivotal for integrin activation, but also directly links integrins to the actin cytoskeleton. Here, we found that mice lacking talin1 specifically in podocytes display severe proteinuria, foot process effacement, and kidney failure. Loss of talin1 in podocytes caused only a modest reduction in β1 integrin activation, podocyte cell adhesion, and cell spreading; however, the actin cytoskeleton of podocytes was profoundly altered by the loss of talin1. Evaluation of murine models of glomerular injury and patients with nephrotic syndrome revealed that calpain-induced talin1 cleavage in podocytes might promote pathogenesis of nephrotic syndrome. Furthermore, pharmacologic inhibition of calpain activity following glomerular injury substantially reduced talin1 cleavage, albuminuria, and foot process effacement. Collectively, these findings indicate that podocyte talin1 is critical for maintaining the integrity of the glomerular filtration barrier and provide insight into the pathogenesis of nephrotic syndrome.
Collapse
Affiliation(s)
- Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Jin Ju Kim
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Susan M. Monkley
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Nanami Gotoh
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ramiro Nandez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Keita Soda
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Daniel M. Balkin
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Hossam Hassan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sung Hyun Son
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Yashang Lee
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Gilbert Moeckel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - David A. Calderwood
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Lawrence B. Holzman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - David R. Critchley
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Roy Zent
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Jochen Reiser
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Miami, Miami, Florida, USA.
Department of Biochemistry, University of Leicester, Leicester, United Kingdom.
Department of Cell Biology,
Howard Hughes Medical Institute,
Program in Cellular Neuroscience, Neurodegeneration, and Repair,
Department of Pathology, and
Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.
Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Department of Medicine, Vanderbilt University and Veterans Affairs Hospital, Nashville, Tennessee, USA.
Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
22
|
Kraemer BF, Weyrich AS, Lindemann S. Protein degradation systems in platelets. Thromb Haemost 2013; 110:920-4. [PMID: 24048267 DOI: 10.1160/th13-03-0183] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/28/2013] [Indexed: 01/05/2023]
Abstract
Protein synthesis and degradation are essential processes that allow cells to survive and adapt to their surrounding milieu. In nucleated cells, the degradation and/or cleavage of proteins is required to eliminate aberrant proteins. Cells also degrade proteins as a mechanism for cell signalling and complex cellular functions. Although the last decade has convincingly shown that platelets synthesise proteins, the roles of protein degradation in these anucleate cytoplasts are less clear. Here we review what is known about protein degradation in platelets placing particular emphasis on the proteasome and the cysteine protease calpain.
Collapse
Affiliation(s)
- B F Kraemer
- Andrew Weyrich, MD, Eccles Institute of Human Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Building 533 Room 4220, Salt Lake City, Utah 84112, USA, Tel: +1 801 5850702, Fax: +1 801 5850701, E-mail:
| | | | | |
Collapse
|
23
|
|
24
|
Miyazaki T, Koya T, Kigawa Y, Oguchi T, Lei XF, Kim-Kaneyama JR, Miyazaki A. Calpain and atherosclerosis. J Atheroscler Thromb 2012; 20:228-37. [PMID: 23171729 DOI: 10.5551/jat.14787] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This review highlights the pro-atherogenic roles of Ca(2+)-sensitive intracellular protease calpains. Among more than ten species of calpain isozymes, µ- and m-calpains have been characterized most extensively. These two isozymes are ubiquitously expressed in mammalian tissues, including blood vessels, and tightly regulate functional molecules in the vascular component cells through limited proteolytic cleavage. Indeed, previous cell-based experiments showed that calpains play significant roles in nitric oxide production in vascular endothelial cells (ECs), maintenance of EC barrier function and angiogenesis for maintaining vascular homeostasis. Recently, we demonstrated that modified-low density lipoprotein (LDL)-induced m-calpain causes hyperpermeability in ECs, leading to the infiltration of monocytes/macrophages and plasma lipids into the intimal spaces (Miyazaki T. et al., Circulation. 2011; 124: 2522-2532). Calpains also mediate oxidized LDL-induced apoptotic death in ECs. In monocytes/macrophages, calpains induce proteolytic degradation of ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1), which results in impaired cholesterol efflux and subsequent macrophage foam cell formation. In vascular smooth muscle cells, calpains may be involved in the conversion from contractile phenotype to proliferative phenotype. In hepatocytes, calpains disrupt the biogenesis of high-density lipoprotein via proteolytic degradation of ABCA1. Thus, calpains may serve as novel candidate molecular targets for control of atherosclerosis.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine, Tokyo 142-8555, Japan.
| | | | | | | | | | | | | |
Collapse
|
25
|
Waldeck H, Wang X, Joyce E, Kao WJ. Active leukocyte detachment and apoptosis/necrosis on PEG hydrogels and the implication in the host inflammatory response. Biomaterials 2011; 33:29-37. [PMID: 21963150 DOI: 10.1016/j.biomaterials.2011.09.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/21/2011] [Indexed: 02/07/2023]
Abstract
Monocytes/Macrophages have long been recognized as key players in inflammation and wound healing and are often employed in vitro to gain an understanding of the inflammatory response to biomaterials. Previous work has demonstrated a drastic decrease in primary monocyte adherent density on biomaterial surfaces coupled with a change in monocyte behavior over time. However, the mechanism responsible for this decrease remains unclear. In this study, we explored active detachment and cellular death as possible regulating factors. Specifically, extracellular TNF-α and ROS production were analyzed as potential endogenous stimulators of cell death. MMPs, but not calpains, were found to play a key role in active monocyte detachment. Monocyte death was found to peak at 24 h and occur by both apoptosis and necrosis as opposed to polymorphonuclear leukocyte death which mainly occurred through apoptosis. Finally, TNF-α and ROS production were not found to have a causal relationship with monocyte death on TCPS or PEG surfaces. The occurrence of primary monocyte apoptosis/necrosis as well as active detachment from a material surface has implications not only in in vitro study, but also in the translation of the in vitro inflammatory response of these cells to in vivo applications.
Collapse
Affiliation(s)
- Heather Waldeck
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, WI, USA.
| | | | | | | |
Collapse
|
26
|
Abstract
The calpains are a conserved family of cysteine proteinases that catalyse the controlled proteolysis of many specific substrates. Calpain activity is implicated in several fundamental physiological processes, including cytoskeletal remodelling, cellular signalling, apoptosis and cell survival. Calpain expression is altered during tumorigenesis, and the proteolysis of numerous substrates, such as inhibitors of nuclear factor-κB (IκB), focal adhesion proteins (including, focal adhesion kinase and talin) and proto-oncogenes (for example, MYC), has been implicated in tumour pathogenesis. Recent evidence indicates that the increased expression of certain family members might influence the response to cancer therapies, providing justification for the development of novel calpain inhibitors.
Collapse
Affiliation(s)
- Sarah J Storr
- University of Nottingham, School of Molecular Medical Sciences, Nottingham NG5 1PB, UK
| | | | | | | | | |
Collapse
|
27
|
Leloup L, Shao H, Bae YH, Deasy B, Stolz D, Roy P, Wells A. m-Calpain activation is regulated by its membrane localization and by its binding to phosphatidylinositol 4,5-bisphosphate. J Biol Chem 2010; 285:33549-33566. [PMID: 20729206 DOI: 10.1074/jbc.m110.123604] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
m-Calpain plays a critical role in cell migration enabling rear de-adhesion of adherent cells by cleaving structural components of the adhesion plaques. Growth factors and chemokines regulate keratinocyte, fibroblast, and endothelial cell migration by modulating m-calpain activity. Growth factor receptors activate m-calpain secondary to phosphorylation on serine 50 by ERK. Concurrently, activated m-calpain is localized to its inner membrane milieu by binding to phosphatidylinositol 4,5-bisphosphate (PIP(2)). Opposing this, CXCR3 ligands inhibit cell migration by blocking m-calpain activity secondary to a PKA-mediated phosphorylation in the C2-like domain. The failure of m-calpain activation in the absence of PIP(2) points to a key regulatory role, although whether this PIP(2)-mediated membrane localization is regulatory for m-calpain activity or merely serves as a docking site for ERK phosphorylation is uncertain. Herein, we report the effects of two CXCR3 ligands, CXCL11/IP-9/I-TAC and CXCL10/IP-10, on the EGF- and VEGF-induced redistribution of m-calpain in human fibroblasts and endothelial cells. The two chemokines block the tail retraction and, thus, the migration within minutes, preventing and reverting growth factor-induced relocalization of m-calpain to the plasma membrane of the cells. PKA phosphorylation of m-calpain blocks the binding of the protease to PIP(2). Unexpectedly, we found that this was due to membrane anchorage itself and not merely serine 50 phosphorylation, as the farnesylation-induced anchorage of m-calpain triggers a strong activation of this protease, leading notably to an increased cell death. Moreover, the ERK and PKA phosphorylations have no effect on this membrane-anchored m-calpain. However, the presence of PIP(2) is still required for the activation of the anchored m-calpain. In conclusion, we describe a novel mechanism of m-calpain activation by interaction with the plasma membrane and PIP(2) specifically, this phosphoinositide acting as a cofactor for the enzyme. The phosphorylation of m-calpain by ERK and PKA by growth factors and chemokines, respectively, act in cells to regulate the enzyme only indirectly by controlling its redistribution.
Collapse
Affiliation(s)
- Ludovic Leloup
- From the Departments of Pathology, Pittsburgh, Pennsylvania 15261
| | - Hanshuang Shao
- From the Departments of Pathology, Pittsburgh, Pennsylvania 15261
| | - Yong Ho Bae
- Bioengineering, Pittsburgh, Pennsylvania 15261
| | | | - Donna Stolz
- Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Partha Roy
- From the Departments of Pathology, Pittsburgh, Pennsylvania 15261; Bioengineering, Pittsburgh, Pennsylvania 15261
| | - Alan Wells
- From the Departments of Pathology, Pittsburgh, Pennsylvania 15261; Bioengineering, Pittsburgh, Pennsylvania 15261; Pittsburgh Veterans Affairs Medical Center, Pittsburgh, Pennsylvania 15261.
| |
Collapse
|
28
|
Carisey A, Ballestrem C. Vinculin, an adapter protein in control of cell adhesion signalling. Eur J Cell Biol 2010; 90:157-63. [PMID: 20655620 PMCID: PMC3526775 DOI: 10.1016/j.ejcb.2010.06.007] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/21/2010] [Accepted: 06/23/2010] [Indexed: 01/09/2023] Open
Abstract
Vinculin, discovered in 1979 (Geiger, 1979), is an adapter protein with binding sites for more than 15 proteins. Biochemical and structural analyses have contributed to detailed knowledge about potential binding partners and the understanding of how their binding may be regulated. Despite all this information the molecular basis of how vinculin acts in cells and controls a wide variety of signals remains elusive. This review aims to highlight recent discoveries with an emphasis on how vinculin is involved in the coordination of a network of signals.
Collapse
Affiliation(s)
- Alex Carisey
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, UK
| | | |
Collapse
|
29
|
Yamada M, Yoshida Y, Mori D, Takitoh T, Kengaku M, Umeshima H, Takao K, Miyakawa T, Sato M, Sorimachi H, Wynshaw-Boris A, Hirotsune S. Inhibition of calpain increases LIS1 expression and partially rescues in vivo phenotypes in a mouse model of lissencephaly. Nat Med 2009; 15:1202-7. [PMID: 19734909 PMCID: PMC2759411 DOI: 10.1038/nm.2023] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 07/30/2009] [Indexed: 12/17/2022]
Abstract
Lissencephaly is a devastating neurological disorder due to defective neuronal migration. LIS1 (or PAFAH1B1) was identified as the gene mutated in lissencephaly patients, and was found to regulate cytoplasmic dynein function and localization. Here, we show that more than half of LIS1 is degraded via calpain-dependent proteolysis, and that inhibition or knockdown of calpains protects LIS1 from proteolysis, resulting in the augmentation of LIS1 levels in Lis1+/− mouse embryonic fibroblast (MEF) cells, which leads to rescue of the aberrant distribution of cytoplasmic dynein, mitochondria and β-COP positive vesicles. We also show that calpain inhibitors improve neuronal migration of Lis1+/− cerebellar granular neurons. Intra-peritoneal injection of ALLN to pregnant Lis1+/− dams rescued apoptotic neuronal cell death and neuronal migration defects in Lis1+/− offspring. Furthermore, in utero knockdown of calpain by shRNA rescued defective cortical layering in Lis1+/− mice. Thus, the inhibition of calpain is a potential therapeutic intervention for lissencephaly.
Collapse
Affiliation(s)
- Masami Yamada
- Department of Genetic Disease Research, Osaka City University Graduate School of Medicine, Asahi-machi, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Seimiya M, Tomonaga T, Matsushita K, Sunaga M, Oh-Ishi M, Kodera Y, Maeda T, Takano S, Togawa A, Yoshitomi H, Otsuka M, Yamamoto M, Nakano M, Miyazaki M, Nomura F. Identification of novel immunohistochemical tumor markers for primary hepatocellular carcinoma; clathrin heavy chain and formiminotransferase cyclodeaminase. Hepatology 2008; 48:519-30. [PMID: 18571811 DOI: 10.1002/hep.22364] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
UNLABELLED Early diagnosis of hepatocellular carcinoma (HCC) greatly improves its prognosis. However, the distinction between benign and malignant tumors is often difficult, and novel immunohistochemical markers are necessary. Using agarose two-dimensional fluorescence difference gel electrophoresis, we analyzed HCC tissues from 10 patients. The fluorescence volumes of 48 spots increased and 79 spots decreased in tumor tissues compared with adjacent nontumor tissue, and 83 proteins were identified by mass spectrometry. Immunoblot confirmed that the expression of clathrin heavy chain (CHC) and Ku86 significantly increased, whereas formiminotransferase cyclodeaminase (FTCD), rhodanese, and vinculin decreased in tumor. The protein expression in tumor and nontumor tissues was further evaluated by immunostaining. Interestingly, CHC and FTCD expression was strikingly different between tumor and nontumor tissues. The sensitivity and specificity of individual markers or a combination for the detection of HCC were 51.8% and 95.6% for CHC, 61.4% and 98.5% for FTCD, and 80.7% and 94.1% for CHC+FTCD, respectively. Strikingly, the sensitivity and specificity increased to 86.7% and 95.6% when glypican-3, another potential biomarker for HCC, was used with FTCD. Moreover, CHC and FTCD were useful to distinguish early HCC from benign tumors such as regenerative nodule or focal nodular hyperplasia, because the sensitivity and specificity of the markers are 41.2% and 77.8% for CHC, 44.4% and 80.0% for FTCD, which is comparable with those of glypican-3 (33.3% and 100%). The sensitivity significantly increased by combination of these markers, 72.2% for CHC+FTCD, and 61.1% for CHC+glypican-3 and FTCD+glypican-3, as 44.4% of glypican-3 negative early HCC were able to be detected by either CHC or FTCD staining. CONCLUSION Immunostaining of CHC and FTCD could make substantial contributions to the early diagnosis of HCC.
Collapse
Affiliation(s)
- Masanori Seimiya
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Goudenege S, Dargelos E, Claverol S, Bonneu M, Cottin P, Poussard S. Comparative proteomic analysis of myotube caveolae after milli-calpain deregulation. Proteomics 2007; 7:3289-98. [PMID: 17849407 DOI: 10.1002/pmic.200700124] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Caveolae are specialised RAFTs (detergent-resistant membrane microdomains enriched in cholesterol and glycosphingolipids). Caveolin, the main caveolae protein, is essential to the organisation of proteins and lipids, and interacts with numerous mediating proteins through a 'Caveolin Scalfolding Domain'. Consequently, caveolae play a major role in signal transduction and appear to be veritable signalling platforms. In muscle cells, caveolae are essential for fusion and differentiation, and are also implicated in a type of muscular dystrophy (LGMD1C). In a preceding work, we demonstrated the presence of active milli-calpain (m-calpain) in myotube caveolae. Calpains are calcium-dependent proteases involved in several cellular processes, including myoblast fusion and migration, PKC-mediated intracellular signalling and remodelling of the cytoskeleton. For the first time, we have proved the cholesterol-dependent localisation of m-calpain in the caveolae of C(2)C(12) myotubes. Calpain-dependent caveolae involvement in myoblast fusion was also strongly suggested. Furthermore, eight differentially expressed caveolae associated proteins were identified by 2-DE and LC-MS/MS analyses using an m-calpain antisense strategy. This proteomic study also demonstrates the action of m-calpain on vimentin, desmin and vinculin in myotube caveolae and suggests m-calpain's role in several mitochondrial pathways.
Collapse
Affiliation(s)
- Sébastien Goudenege
- Université Bordeaux I, USC-INRA 2009, Unité Protéolyse, Croissance et Développement Musculaire, ISTAB, Talence, France
| | | | | | | | | | | |
Collapse
|
32
|
Dargelos E, Poussard S, Brulé C, Daury L, Cottin P. Calcium-dependent proteolytic system and muscle dysfunctions: a possible role of calpains in sarcopenia. Biochimie 2007; 90:359-68. [PMID: 17881114 DOI: 10.1016/j.biochi.2007.07.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Accepted: 07/20/2007] [Indexed: 02/02/2023]
Abstract
The calcium-dependent proteolytic system is composed of cysteine proteases named calpains. They are ubiquitous or tissue-specific enzymes. The two best characterised isoforms are the ubiquitously expressed mu- and m-calpains. Besides its regulation by calcium, calpain activity is tightly controlled by calpastatin, the specific endogenous inhibitor, binding to phospholipids, autoproteolysis and phosphorylation. Calpains are responsible for limited proteolytic events. Among the multitude of substrates identified so far are cytoskeletal and membrane proteins, enzymes and transcription factors. Calpain activity is involved in a large number of physiological and pathological processes. In this review, we will particularly focus on the implication of the calcium-dependent proteolytic system in relation to muscle physiology. Because of their ability to remodel cytoskeletal anchorage complexes, calpains play a major role in the regulation of cell adhesion, migration and fusion, three key steps of myogenesis. Calcium-dependent proteolysis is also involved in the control of cell cycle. In muscle tissue, in particular, calpains intervene in the regeneration process. Another important class of calpain substrates belongs to apoptosis regulating factors. The proteases may thus play a role in muscle cell death, and as a consequence in muscle atrophy. The relationships between calcium-dependent proteolysis and muscle dysfunctions are being further developed in this review with a particular emphasis on sarcopenia.
Collapse
Affiliation(s)
- E Dargelos
- Université Bordeaux I, INRA USC-2009, Unité Protéolyse Croissance et Développement Musculaire, ISTAB, avenue des facultés, 33405 Talence cedex, France.
| | | | | | | | | |
Collapse
|
33
|
Miyazaki T, Honda K, Ohata H. Requirement of Ca2+ influx- and phosphatidylinositol 3-kinase-mediated m-calpain activity for shear stress-induced endothelial cell polarity. Am J Physiol Cell Physiol 2007; 293:C1216-25. [PMID: 17596297 DOI: 10.1152/ajpcell.00083.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteolytic activity in sheared human umbilical vein endothelial cells (HUVECs) was measured using a fluorogenic substrate and laser scanning confocal microscopy to clarify the key role of an intracellular Ca(2+)-sensitive protease, calpain, in these cells in response to shear stress. Within physiological shear range, activity in the cells was enhanced in shear-dependent fashion. Short interfering RNA-induced silencing of m-calpain, but not of micro-calpain, suppressed the activity. Either removal of extracellular Ca(2+) or application of an intracellular Ca(2+) chelator (BAPTA/AM) or nonselective cation channel blocker (Gd(3+)) reduced proteolytic activity. Furthermore, activity was suppressed by phosphatidylinositol bisphosphate (PIP(2)) chelator (neomycin) or phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002); in contrast, activity, which was partially inhibited by ERK kinase inhibitor (U0126, PD98059), was unaffected by PLC inhibitor (U73122). Moreover, Akt phosphorylation downstream of PI3K, which was elicited by shear, was attenuated by neomycin but not by calpain inhibitor (calpeptin). Following assessment of shear stress-induced focal adhesion (FA) and cytoskeletal dynamics using interference reflection/green fluorescence protein-actin microscopy, we found that either calpain or PI3K inhibition impaired shear stress-induced polarization of FAs via stabilization of FA structures. Additionally, HUVEC alignment and cytoskeletal remodeling, which was accompanied by calpain-mediated cleavage of vinculin and talin, were also elicited by prolonged application of shear and impaired by m-calpain knockdown. Thus, these results revealed that physiological shear stress elicits Ca(2+) influx-sensitive activation of m-calpain in HUVECs. This activity is facilitated primarily through the PI3K pathway; furthermore, it is essential for subsequent FA reorganization and cell alignment under shear conditions.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Pharmacology, School of Pharmaceutical Sciences, Showa University, Tokyo, Japan.
| | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW There is considerable interest in understanding the function and mechanism of calpains in platelet aggregation, spreading, and granular secretion pathways. Recent insights from the calpain-1 knockout platelets suggest a pivotal role of these cysteine proteases in the regulation of outside-in signaling, aggregation, and clot retraction. RECENT FINDINGS The calpain-1 knockout mouse provided direct evidence for the role of calpain-1 in platelet aggregation and clot retraction. Reduced tyrosine phosphorylation of platelet proteins correlated with reduced platelet aggregation and clot retraction. Future investigations of the mechanism of platelet defects in calpain-1 null mice may unveil the physiological functions of this important and elusive protease in mammalian cells. SUMMARY This review focuses on the role of calpains in platelets with a particular emphasis on recent findings in calpain-1 null platelets. Previous studies used synthetic inhibitors to study the role of calpains in platelet function yielding useful information about the identification of calpain substrates. The development of calpain-1 null mice demonstrated that calpain-1 plays an important function in the regulation of platelet aggregation and clot retraction. Since the combined deletion of calpain-1 and calpain-2 genes results in embryonic lethality, the calpain-1 null mouse remains the only experimental model available to study the physiological role of calpains in mammalian cells.
Collapse
Affiliation(s)
- Shafi M Kuchay
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | |
Collapse
|
35
|
Abstract
Cells offer different types of cytoskeletal anchorages: transitory structures such as focal contacts and perennial ones such as the sarcomeric cytoskeleton of muscle cells. The turnover of these structures is controlled with different timing by a family of cysteine proteases activated by calcium, the calpains. The large number of potential substrates present in each of these structures imposes fine tuning of the activity of the proteases to avoid excessive action. This phenomenon is thus guaranteed by various types of regulation, ranging from a relatively high calcium concentration necessary for activation, phosphorylation of substrates or the proteases themselves with either a favorable or inhibitory effect, possible intervention of phospholipids, and the presence of a specific inhibitor and its possible degradation before activation. Finally, formation of multiprotein complexes containing calpains offers a new method of regulation.
Collapse
|
36
|
Mazères G, Leloup L, Daury L, Cottin P, Brustis JJ. Myoblast attachment and spreading are regulated by different patterns by ubiquitous calpains. ACTA ACUST UNITED AC 2006; 63:193-207. [PMID: 16496301 DOI: 10.1002/cm.20116] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The calcium-dependent proteolytic system is a large family of well-conserved ubiquitous and tissue-specific proteases, known as calpains, and an endogenous inhibitor, calpastatin. Ubiquitous calpains are involved in many physiological phenomena, such as the cell cycle, muscle cell differentiation, and cell migration. This study investigates the regulation of crucial steps of cell motility, myoblast adhesion and spreading, by calpains. Inhibition of each ubiquitous calpain isoform by antisense strategy pinpointed the involvement of each of these proteases in myoblast adhesion and spreading. Moreover, the actin cytoskeleton and microtubules were observed in transfected cells, demonstrating that each ubiquitous calpain could be involved in the actin fiber organization. C2C12 cells with reduced mu- or m-calpain levels have a rounded morphology and disorganized stress fibers, but no modification in the microtubule cytoskeleton. Antisense strategy directed against MARCKS, a calpain substrate during C2C12 migration, showed that this protein could play a role in stress fiber polymerization. A complementary proteomic analysis using C2C12 cells over-expressing calpastatin indicated that two proteins were under-expressed, while six, which are involved in the studied phenomena, were overexpressed after calpain inhibition. The possible role of these proteins in adhesion, spreading, and migration was discussed.
Collapse
Affiliation(s)
- Germain Mazères
- Laboratoire Biosciences de l'Aliment, ISTAB USC-INRA 2009, Talence Cedex, France
| | | | | | | | | |
Collapse
|
37
|
Bachelot-Loza C, Badol P, Brohard-Bohn B, Fraiz N, Cano E, Rendu F. Differential regulation of platelet aggregation and aminophospholipid exposure by calpain. Br J Haematol 2006; 133:419-26. [PMID: 16643450 DOI: 10.1111/j.1365-2141.2006.06031.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aggregation, exposure of procoagulant phospholipids and shedding of microparticles are platelet responses that depend on activating conditions. To determine how these different responses are interconnected, we simultaneously measured fibrinogen (Fg) binding and aminophospholipid exposure on activated platelets by means of flow cytometry. Low calcium ionophore (A23187) concentrations induced Fg binding but not annexin V binding. In contrast, high A23187 concentrations induced annexin V binding but not Fg binding. Collagen, both alone and in the presence of thrombin, induced both Fg and annexin V binding. Dual labelling was found on 38 +/- 9% of platelets stimulated by thrombin plus collagen. The regulatory role of calpain in these platelet functions was investigated. When calpain was partially inhibited by 2 microg/ml calpeptin, Fg binding still occurred but aminophospholipid exposure was limited. By contrast, complete inhibition of calpain by 100 microg/ml calpeptin or E64d decreased Fg binding but enhanced aminophospholipid exposure. In these latter conditions, cytosolic calcium-extruding systems were inhibited. The results suggest that (i) conditions that favour aminophospholipid exposure tend to decrease the aggregation process and (ii) calpain determines the switch to either aggregation or aminophospholipid exposure by controlling intracellular calcium.
Collapse
Affiliation(s)
- Christilla Bachelot-Loza
- INSERM U765, Faculté des Sciences Pharmaceutiques et biologiques, Université Paris V, Paris, France.
| | | | | | | | | | | |
Collapse
|
38
|
Cathelin S, Rébé C, Haddaoui L, Simioni N, Verdier F, Fontenay M, Launay S, Mayeux P, Solary E. Identification of proteins cleaved downstream of caspase activation in monocytes undergoing macrophage differentiation. J Biol Chem 2006; 281:17779-88. [PMID: 16636047 DOI: 10.1074/jbc.m600537200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We have shown previously that caspases were specifically involved in the differentiation of peripheral blood monocytes into macrophages while not required for monocyte differentiation into dendritic cells. To identify caspase targets in monocytes undergoing macrophagic differentiation, we used the human monocytic leukemic cell line U937, whose macrophagic differentiation induced by exposure to 12-O-tetradecanoylphorbol 13-acetate (TPA) can be prevented by expression of the baculovirus caspase-inhibitory protein p35. A comparative two-dimensional gel proteomic analysis of empty vector- and p35-transfected cells after 12 h of exposure to 20 nm TPA, followed by mass spectrometry analysis, identified 38 differentially expressed proteins. Those overexpressed in p35-expressing cells (n = 16) were all full-length, whereas half of those overexpressed in control cells (n = 22) were N- or C-terminal cleavage fragments. The cleavage or degradation of seven of these proteins was confirmed in peripheral blood monocytes undergoing macrophage colony-stimulating factor-induced macrophagic differentiation. In U937 cells exposed to TPA, these proteolytic events can be inhibited by expression of a caspase-8 dominant negative mutant or the cowpox virus CrmA caspase inhibitor. These cleavages provide new insights to analyze the role of caspases in this specific differentiation program.
Collapse
Affiliation(s)
- Séverine Cathelin
- INSERM UMR 517, IFR 100, Faculty of Medicine, 7 Boulevard Jeanne d'Arc, F-21079 Dijon Cedex, France
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The calpain family of proteases has been implicated in cellular processes such as apoptosis, proliferation and cell migration. Calpains are involved in several key aspects of migration, including: adhesion and spreading; detachment of the rear; integrin- and growth-factor-mediated signaling; and membrane protrusion. Our understanding of how calpains are activated and regulated during cell migration has increased as studies have identified roles for calcium and phospholipid binding, autolysis, phosphorylation and inhibition by calpastatin in the modulation of calpain activity. Knockout and knockdown approaches have also contributed significantly to our knowledge of calpain biology, particularly with respect to the specific functions of different calpain isoforms. The mechanisms by which calpain-mediated proteolysis of individual substrates contributes to cell motility have begun to be addressed, and these efforts have revealed roles for proteolysis of specific substrates in integrin activation, adhesion complex turnover and membrane protrusion dynamics. Understanding these mechanisms should provide avenues for novel therapeutic strategies to treat pathological processes such as tumor metastasis and chronic inflammatory disease.
Collapse
Affiliation(s)
- Santos J Franco
- Program in Cellular and Molecular Biology, University of Wisconsin, Madison, WI 53705, USA
| | | |
Collapse
|
40
|
Miyazaki T, Yamamoto M, Honda K, Ohata H. [Flow-induced Ca2+ transients regulate morphological changes in vascular endothelial cells via calpain-mediated proteolytic cleavage]. Nihon Yakurigaku Zasshi 2005; 126:256-61. [PMID: 16327206 DOI: 10.1254/fpj.126.256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
41
|
Lavastre V, Chiasson S, Cavalli H, Girard D. Viscum album agglutinin-I (VAA-I) induces apoptosis and degradation of cytoskeletal proteins in human leukemia PLB-985 and X-CGD cells via caspases: Lamin B1 is a novel target of VAA-I. Leuk Res 2005; 29:1443-53. [PMID: 16242777 DOI: 10.1016/j.leukres.2005.05.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2005] [Accepted: 05/12/2005] [Indexed: 01/12/2023]
Abstract
Viscum album agglutinin-I (VAA-I) is a potent inducer of cell apoptosis and possesses anti-tumoral activity. Using PLB-985 and chronic granulomatous disease (X-CGD) cells, which lack expression of gp91(phox), VAA-I was found to induce apoptosis in both cell lines as assessed by cytology, DNA laddering and degradation of the cytoskeletal protein gelsolin. Both cell lines expressed caspase-3 and -8 and VAA-I activated these caspases. We demonstrated that lamin B(1) is a novel target to VAA-I and its degradation was reversed by a pan-caspase inhibitor and by a caspase-6, but not a caspase-8, inhibitor.
Collapse
Affiliation(s)
- Valérie Lavastre
- Institut National de Recherche Scientifique, INRS-Institut Armand-Frappier, Pointe-Claire, Canada
| | | | | | | |
Collapse
|