1
|
Abady MM, Saadeldin IM, Han A, Bang S, Kang H, Seok DW, Kwon HJ, Cho J, Jeong JS. Melatonin and resveratrol alleviate molecular and metabolic toxicity induced by Bisphenol A in endometrial organoids. Reprod Toxicol 2024; 128:108628. [PMID: 38848930 DOI: 10.1016/j.reprotox.2024.108628] [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: 04/02/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
Bisphenol A (BPA), a widespread environmental contaminant, poses concerns due to its disruptive effects on physiological functions of the uterine endometrium. In contrast, melatonin (MT) and Resveratrol (RSV) are under scrutiny for their potential protective roles against BPA-induced damage. For the efficacy and ethical concerns in the animal test, endometrial organoids, three-dimensional models mimicking endometrium, serve as crucial tools for unraveling the impact of environmental factors on reproductive health. This study aimed to comprehensively characterize the morphological, molecular and metabolic responses of porcine endometrial organoids to BPA and assess the potential protective effects of MT and RSV. Porcine uteri were prepared, digested with collagenase, mixed with Matrigel, and incubated at 38°C with 5 % CO2. Passaging involved dissociation through trypsin-EDTA treatment and subculturing. The culture medium was refreshed every 2-3 days. To investigate the environmental impact on reproductive health, endometrial organoids were treated with BPA (0.5 µM), MT (with/without BPA at 0.1 µM), and/or RSV (10 µM). Various molecular screening using gene expression, western blotting, immunofluorescence staining, and metabolites profiling were assessed the effects of BPA, MT, and RSV in terms of cell viability, morphology, reproductivity, and metabolism alteration in the endometrial organoids. As expected, BPA induced structural and molecular disruptions in organoids, affecting cytoskeletal proteins, Wnt/β-catenin signaling, and epithelial/mesenchymal markers. It triggered oxidative stress and apoptotic pathways, altered miRNA expression, and disrupted the endocannabinoid system. The level of glucose, galactose, and essential amino acids were increased or decreased by approximately 1.5-3 times in BPA-treated groups compared to the control groups (p-value < 0.05), indicating metabolic changes. Moreover, MT and RSV treated groups exhibited protective effects, mitigating BPA-induced disruptions across multiple pathways. For the first time, our study models endometrial organoids, advancing understanding of environmental impacts on reproductive health.
Collapse
Affiliation(s)
- Mariam M Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Nutrition and Food Science, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Islam M Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Ayeong Han
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seonggyu Bang
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Heejae Kang
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dong Wook Seok
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
2
|
Coelho-Rato LS, Parvanian S, Andrs Salajkova S, Medalia O, Eriksson JE. Intermediate filaments at a glance. J Cell Sci 2024; 137:jcs261386. [PMID: 39206824 DOI: 10.1242/jcs.261386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Intermediate filaments (IFs) comprise a large family of versatile cytoskeletal proteins, divided into six subtypes with tissue-specific expression patterns. IFs have a wide repertoire of cellular functions, including providing structural support to cells, as well as active roles in mechanical support and signaling pathways. Consequently, defects in IFs are associated with more than 100 diseases. In this Cell Science at a Glance article, we discuss the established classes of IFs and their general features, their functions beyond structural support, and recent advances in the field. We also highlight their involvement in disease and potential use as clinical markers of pathological conditions. Finally, we provide our view on current knowledge gaps and the future directions of the IF field.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Sarka Andrs Salajkova
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Euro-Bioimaging ERIC, 20520 Turku, Finland
| |
Collapse
|
3
|
Fragkiadakis K, Ktena N, Kalantidou A, Dermitzaki E, Anastasiou I, Papathanassiou S, Kontaraki J, Kalomoirakis P, Kanoupakis E, Patrianakos A, Papadomanolakis A, Daskalaki E, Kiousi T, Kouraki K, Kranioti E, Tzardi M, Venihaki M, Karagogeos D, Capetanaki Y, Kardassis D, Kochiadakis G, Parthenakis F, Marketou M. Cytokeratin 18 as a Novel Biomarker in Patients with Hypertrophic Cardiomyopathy. Cells 2024; 13:1328. [PMID: 39195218 PMCID: PMC11352956 DOI: 10.3390/cells13161328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a heart muscle disease associated with an increased risk for sudden cardiac death (SCD). Cytokeratin 18-based proteins, such as M30 and M65 antigens, are known cell-death biomarkers. M30 antigen is released from cells during apoptosis, and M65 antigen is released during cell death from any cause, such as apoptosis or necrosis. We aimed to study the expression of M30 and M65 antigens in peripheral blood obtained by 46 HCM patients and compare with 27 age- and sex-matched patients without HCM. We also investigated the CK18 expression in myocardium from postmortem HCM hearts. M30 and M65 antigens were significantly increased in the HCM vs. non-HCM group (Μ30: 338 ± 197 U/uL vs. 206 ± 166 U/uL, p = 0.003; M65: 428 ± 224 U/uL vs. 246 ± 214 U/uL, p = 0.001), and HCM patients with a higher expression of these markers (M30: 417 ± 208 vs. 271 ± 162 U/uL, p = 0.011; M65: 518 ± 242 vs. 351 ± 178 U/uL, p = 0.011) had a higher risk for SCD. In HCM, both apoptosis and necrosis are increased, but particularly necrosis (M30/M65 ratio: 0.75 ± 0.09 vs. 0.85 ± 0.02, p < 0.001). CK18 is expressed in the HCM myocardium (1.767 ± 0.412 vs. 0.537 ± 0.383, % of area, p = 0.0058). Therefore, M30 and M65 antigens may be novel biomarkers in HCM.
Collapse
Affiliation(s)
- Konstantinos Fragkiadakis
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
- Cardiology Department, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Niki Ktena
- Division of Basic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece (D.K.)
| | - Aikaterini Kalantidou
- Clinical Chemistry, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.K.); (M.V.)
| | - Eirini Dermitzaki
- Clinical Chemistry, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.K.); (M.V.)
| | - Ioannis Anastasiou
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
| | | | - Joanna Kontaraki
- Cardiology Department, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Petros Kalomoirakis
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
| | - Emmanuel Kanoupakis
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
| | - Alexandros Patrianakos
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
| | - Antonis Papadomanolakis
- Forensic Medicine Unit, Department of Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Efsevia Daskalaki
- Forensic Medicine Unit, Department of Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Theodora Kiousi
- Forensic Medicine Unit, Department of Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Katerina Kouraki
- Laboratory of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Greece
| | - Elena Kranioti
- Forensic Medicine Unit, Department of Forensic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Maria Tzardi
- Laboratory of Pathology, University General Hospital of Heraklion, 71110 Heraklion, Greece
| | - Maria Venihaki
- Clinical Chemistry, School of Medicine, University of Crete, 71003 Heraklion, Greece; (A.K.); (M.V.)
| | - Domna Karagogeos
- Division of Basic Sciences, School of Medicine, University of Crete, 71003 Heraklion, Greece (D.K.)
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Georgios Kochiadakis
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
- Cardiology Department, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - Fragkiskos Parthenakis
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
| | - Maria Marketou
- Cardiology Department, Heraklion University General Hospital, 71110 Heraklion, Greece (E.K.); (M.M.)
- Cardiology Department, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| |
Collapse
|
4
|
Ivanov MV, Kopeykina AS, Gorshkov MV. Reanalysis of DIA Data Demonstrates the Capabilities of MS/MS-Free Proteomics to Reveal New Biological Insights in Disease-Related Samples. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1775-1785. [PMID: 38938158 DOI: 10.1021/jasms.4c00134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Data-independent acquisition (DIA) at the shortened data acquisition time is becoming a method of choice for quantitative proteomic applications requiring high throughput analysis of large cohorts of samples. With the advent of the combination of high resolution mass spectrometry with an asymmetric track lossless analyzer, these DIA capabilities were further extended with the recent demonstration of quantitative analyses at the speed of up to hundreds of samples per day. In particular, the proteomic data for the brain samples related to multiple system atrophy disease were acquired using 7 and 28 min chromatography gradients (Guzman et al., Nat. Biotech. 2024). In this work, we applied the recently introduced DirectMS1 method to reanalysis of these data using only MS1 spectra. Both DirectMS1 and DIA results were matched against long gradient DDA analysis from the earlier study of the same sample cohort. While the quantitation efficiency of DirectMS1 was comparable with DIA on the same data sets, we found an additional five proteins of biological significance relevant to the analyzed tissue samples. Among the findings, DirectMS1 was able to detect decreased caspase activity for Vimentin protein in the multiple system atrophy samples missed by the MS/MS-based quantitation methods. Our study suggests that DirectMS1 can be an efficient MS1-only addition to the analysis of DIA data in high-throughput quantitative proteomic studies.
Collapse
Affiliation(s)
- Mark V Ivanov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Anna S Kopeykina
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Mikhail V Gorshkov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center of Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
5
|
Svandova E, Vesela B, Janeckova E, Chai Y, Matalova E. Exploring caspase functions in mouse models. Apoptosis 2024; 29:938-966. [PMID: 38824481 PMCID: PMC11263464 DOI: 10.1007/s10495-024-01976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
Caspases are enzymes with protease activity. Despite being known for more than three decades, caspase investigation still yields surprising and fascinating information. Initially associated with cell death and inflammation, their functions have gradually been revealed to extend beyond, targeting pathways such as cell proliferation, migration, and differentiation. These processes are also associated with disease mechanisms, positioning caspases as potential targets for numerous pathologies including inflammatory, neurological, metabolic, or oncological conditions. While in vitro studies play a crucial role in elucidating molecular pathways, they lack the context of the body's complexity. Therefore, laboratory animals are an indispensable part of successfully understanding and applying caspase networks. This paper aims to summarize and discuss recent knowledge, understanding, and challenges in caspase knock-out mice.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic.
| | - Barbora Vesela
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
| | - Eva Janeckova
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, USA
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetic, Brno, Czech Republic
- Department of Physiology, University of Veterinary Sciences, Brno, Czech Republic
| |
Collapse
|
6
|
Nguyen A, Mustafa AHM, Leydecker AK, Halilovic M, Murr J, Butter F, Krämer OH. The protein phosphatase-2A subunit PR130 is involved in the formation of cytotoxic protein aggregates in pancreatic ductal adenocarcinoma cells. Cell Commun Signal 2024; 22:217. [PMID: 38570831 PMCID: PMC10993613 DOI: 10.1186/s12964-024-01597-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
As a major source of cellular serine and threonine phosphatase activity, protein phosphatase-2A (PP2A) modulates signaling pathways in health and disease. PP2A complexes consist of catalytic, scaffolding, and B-type subunits. Seventeen PP2A B-type subunits direct PP2A complexes to selected substrates. It is ill-defined how PP2A B-type subunits determine the growth and drug responsiveness of tumor cells. Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis. We analyzed the responses of murine and human mesenchymal and epithelial PDAC cells to the specific PP2A inhibitor phendione. We assessed protein levels by immunoblot and proteomics and cell fate by flow cytometry, confocal microscopy, and genetic manipulation. We show that murine mesenchymal PDAC cells express significantly higher levels of the PP2A B-type subunit PR130 than epithelial PDAC cells. This overexpression of PR130 is associated with a dependency of such metastasis-prone cells on the catalytic activity of PP2A. Phendione induces apoptosis and an accumulation of cytotoxic protein aggregates in murine mesenchymal and human PDAC cells. These processes occur independently of the frequently mutated tumor suppressor p53. Proteomic analyses reveal that phendione upregulates the chaperone HSP70 in mesenchymal PDAC cells. Inhibition of HSP70 promotes phendione-induced apoptosis and phendione promotes a proteasomal degradation of PR130. Genetic elimination of PR130 sensitizes murine and human PDAC cells to phendione-induced apoptosis and protein aggregate formation. These data suggest that the PP2A-PR130 complex dephosphorylates and thereby prevents the aggregation of proteins in tumor cells.
Collapse
Affiliation(s)
- Alexandra Nguyen
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Al-Hassan M Mustafa
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
- Department of Zoology, Faculty of Science, Aswan University, Aswan, Egypt
| | - Alessa K Leydecker
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Melisa Halilovic
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany
| | - Janine Murr
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675, Munich, Germany
| | - Falk Butter
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128, Mainz, Germany
- Federal Research Institute for Animal Health, Südufer 10, 17493, Greifswald, Insel Riems, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Obere Zahlbacher St. 67, 55131, Mainz, Germany.
| |
Collapse
|
7
|
Suprewicz Ł, Zakrzewska M, Okła S, Głuszek K, Sadzyńska A, Deptuła P, Fiedoruk K, Bucki R. Extracellular vimentin as a modulator of the immune response and an important player during infectious diseases. Immunol Cell Biol 2024; 102:167-178. [PMID: 38211939 DOI: 10.1111/imcb.12721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024]
Abstract
Vimentin, an intermediate filament protein primarily recognized for its intracellular role in maintaining cellular structure, has recently garnered increased attention and emerged as a pivotal extracellular player in immune regulation and host-pathogen interactions. While the functions of extracellular vimentin were initially overshadowed by its cytoskeletal role, accumulating evidence now highlights its significance in diverse physiological and pathological events. This review explores the multifaceted role of extracellular vimentin in modulating immune responses and orchestrating interactions between host cells and pathogens. It delves into the mechanisms underlying vimentin's release into the extracellular milieu, elucidating its unconventional secretion pathways and identifying critical molecular triggers. In addition, the future perspectives of using extracellular vimentin in diagnostics and as a target protein in the treatment of diseases are discussed.
Collapse
Affiliation(s)
- Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Magdalena Zakrzewska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Sławomir Okła
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Katarzyna Głuszek
- Institute of Medical Sciences, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Alicja Sadzyńska
- State Higher Vocational School of Prof. Edward F. Szczepanik in Suwałki, Suwałki, Poland
| | - Piotr Deptuła
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Białystok, Poland
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
8
|
Yi YS. Roles of the Caspase-11 Non-Canonical Inflammasome in Rheumatic Diseases. Int J Mol Sci 2024; 25:2091. [PMID: 38396768 PMCID: PMC10888639 DOI: 10.3390/ijms25042091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammasomes are intracellular multiprotein complexes that activate inflammatory signaling pathways. Inflammasomes comprise two major classes: canonical inflammasomes, which were discovered first and are activated in response to a variety of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), and non-canonical inflammasomes, which were discovered recently and are only activated in response to intracellular lipopolysaccharide (LPS). Although a larger number of studies have successfully demonstrated that canonical inflammasomes, particularly the NLRP3 inflammasome, play roles in various rheumatic diseases, including rheumatoid arthritis (RA), infectious arthritis (IR), gouty arthritis (GA), osteoarthritis (OA), systemic lupus erythematosus (SLE), psoriatic arthritis (PA), ankylosing spondylitis (AS), and Sjögren's syndrome (SjS), the regulatory roles of non-canonical inflammasomes, such as mouse caspase-11 and human caspase-4 non-canonical inflammasomes, in these diseases are still largely unknown. Interestingly, an increasing number of studies have reported possible roles for non-canonical inflammasomes in the pathogenesis of various mouse models of rheumatic disease. This review comprehensively summarizes and discusses recent emerging studies demonstrating the regulatory roles of non-canonical inflammasomes, particularly focusing on the caspase-11 non-canonical inflammasome, in the pathogenesis and progression of various types of rheumatic diseases and provides new insights into strategies for developing potential therapeutics to prevent and treat rheumatic diseases as well as associated diseases by targeting non-canonical inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
9
|
Cui T, Liu P, Chen X, Liu Z, Wang B, Gao C, Wang Z, Li C, Yang N. Identification and functional characterization of caspases in turbot (Scophthalmus maximus) in response to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108757. [PMID: 37084854 DOI: 10.1016/j.fsi.2023.108757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Apoptosis is the autonomous and orderly death of cells under genetic control to maintain the stability of the internal environment, and is a programmed cell death process with unique morphological and biochemical properties that is regulated by a variety of factors. Caspase gene family has a significant function in the process of apoptosis. However, the knowledge of caspases in turbot remains largely unknown. In present study, a total of nine turbot caspase genes were identified. The mRNA length of these caspase genes was ranged from 1149 bp (caspase-1) to 3216 bp (caspase-2), and the protein length was ranged from 281 aa (caspase-3a) to 507 aa (caspase-10). Phylogenetic analysis showed these caspase genes were divided into three subfamilies. The qRT-PCR results showed that turbot caspase genes were expressed in all the examined organs, especially the intestine, kidney, blood and gills. Meanwhile, we explored the expression patterns of caspase genes in the intestine, skin and gills after Vibrio anguillarum and Aeromonas salmonids infections. The results showed that caspase genes showed different expression patterns in mucosal tissues after bacterial infection, demonstrating the critical role of caspase genes in mucosal immune responses. In addition, protein-protein interaction analysis showed that caspase proteins interacted with immune molecules such as NLR, IL-1β, and birc. The results of interference and overexpression experiments showed that caspase-1 might play key roles in the regulation of the IL-1β production, but the detailed mechanism needs to be further studied. The results of this study provide valuable information for further study the roles of caspase genes in turbot, which could help us to further understand the inflammatory pathways in teleost.
Collapse
Affiliation(s)
- Tong Cui
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Peng Liu
- Yantai Marine Economic Research Institute, Yantai, China
| | - Xuan Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhe Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Beibei Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chengbin Gao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhongyi Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Ning Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
10
|
Barnard SJ, Haunschild J, Heiser L, Dieterlen MT, Klaeske K, Borger MA, Etz CD. Apoptotic Cell Death in Bicuspid-Aortic-Valve-Associated Aortopathy. Int J Mol Sci 2023; 24:ijms24087429. [PMID: 37108591 PMCID: PMC10138609 DOI: 10.3390/ijms24087429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The bicuspid aortic valve (BAV) is the most common cardiovascular congenital abnormality and is frequently associated with proximal aortopathy. We analyzed the tissues of patients with bicuspid and tricuspid aortic valve (TAV) regarding the protein expression of the receptor for advanced glycation products (RAGE) and its ligands, the advanced glycation end products (AGE), as well as the S100 calcium-binding protein A6 (S100A6). Since S100A6 overexpression attenuates cardiomyocyte apoptosis, we investigated the diverse pathways of apoptosis and autophagic cell death in the human ascending aortic specimen of 57 and 49 patients with BAV and TAV morphology, respectively, to identify differences and explanations for the higher risk of patients with BAV for severe cardiovascular diseases. We found significantly increased levels of RAGE, AGE and S100A6 in the aortic tissue of bicuspid patients which may promote apoptosis via the upregulation of caspase-3 activity. Although increased caspase-3 activity was not detected in BAV patients, increased protein expression of the 48 kDa fragment of vimentin was detected. mTOR as a downstream protein of Akt was significantly higher in patients with BAV, whereas Bcl-2 was increased in patients with TAV, assuming a better protection against apoptosis. The autophagy-related proteins p62 and ERK1/2 were increased in patients with BAV, assuming that cells in bicuspid tissue are more likely to undergo apoptotic cell death leading to changes in the wall and finally to aortopathies. We provide first-hand evidence of increased apoptotic cell death in the aortic tissue of BAV patients which may thus provide an explanation for the increased risk of structural aortic wall deficiency possibly underlying aortic aneurysm formation or acute dissection.
Collapse
Affiliation(s)
- Sarah J Barnard
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Josephina Haunschild
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Linda Heiser
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Maja T Dieterlen
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Kristin Klaeske
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Michael A Borger
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| | - Christian D Etz
- Heisenberg Working Group, Saxonian Incubator for Clinical Translation, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
- University Department for Cardiac Surgery, Heart Center Leipzig, 04289 Leipzig, Germany
| |
Collapse
|
11
|
Talukdar SN, McGregor B, Osan JK, Hur J, Mehedi M. RSV infection does not induce EMT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532506. [PMID: 36993657 PMCID: PMC10055011 DOI: 10.1101/2023.03.13.532506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Respiratory syncytial virus (RSV) infection does not cause severe disease in most of us despite suffering from multiple RSV infections in our lives. However, infants, young children, older adults, and immunocompromised patients are unfortunately vulnerable to RSV-associated severe diseases. A recent study suggested that RSV infection causes cell expansion, resulting in bronchial wall thickening in vitro. Whether the virus-induced changes in the lung airway resemble epithelial-mesenchymal transition (EMT) is still unknown. Here, we report that RSV does not induce EMT in three different in vitro lung models: the epithelial A549 cell line, primary normal human bronchial epithelial cells, and pseudostratified airway epithelium. We found that RSV increases the cell surface area and perimeter in the infected airway epithelium, which is distinct from the effects of a potent EMT inducer, TGF-β1-driven cell elongation-indicative of cell motility. A genome-wide transcriptome analysis revealed that both RSV and TGF-β1 have distinct modulation patterns of the transcriptome, which suggests that RSV-induced changes are distinct from EMT.
Collapse
Affiliation(s)
- Sattya N. Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Brett McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Jaspreet K. Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, North Dakota, United States of America
| |
Collapse
|
12
|
Sacristán-Gómez P, Serrano-Somavilla A, Castro-Espadas L, Sánchez de la Blanca Carrero N, Sampedro-Núñez M, Muñoz-De-Nova JL, Molina-Jiménez F, Rosell A, Marazuela M, Martínez-Hernández R. Evaluation of Epithelial-Mesenchymal Transition Markers in Autoimmune Thyroid Diseases. Int J Mol Sci 2023; 24:3359. [PMID: 36834770 PMCID: PMC9965822 DOI: 10.3390/ijms24043359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
A state of chronic inflammation is common in organs affected by autoimmune disorders, such as autoimmune thyroid diseases (AITD). Epithelial cells, such as thyroid follicular cells (TFCs), can experience a total or partial transition to a mesenchymal phenotype under these conditions. One of the major cytokines involved in this phenomenon is transforming growth factor beta (TGF-β), which, at the initial stages of autoimmune disorders, plays an immunosuppressive role. However, at chronic stages, TGF- β contributes to fibrosis and/or transition to mesenchymal phenotypes. The importance of primary cilia (PC) has grown in recent decades as they have been shown to play a key role in cell signaling and maintaining cell structure and function as mechanoreceptors. Deficiencies of PC can trigger epithelial-mesenchymal transition (EMT) and exacerbate autoimmune diseases. A set of EMT markers (E-cadherin, vimentin, α-SMA, and fibronectin) were evaluated in thyroid tissues from AITD patients and controls through RT-qPCR, immunohistochemistry (IHC), and western blot (WB). We established an in vitro TGF-β-stimulation assay in a human thyroid cell line to assess EMT and PC disruption. EMT markers were evaluated in this model using RT-qPCR and WB, and PC was evaluated with a time-course immunofluorescence assay. We found an increased expression of the mesenchymal markers α-SMA and fibronectin in TFCs in the thyroid glands of AITD patients. Furthermore, E-cadherin expression was maintained in these patients compared to the controls. The TGF-β-stimulation assay showed an increase in EMT markers, including vimentin, α-SMA, and fibronectin in thyroid cells, as well as a disruption of PC. The TFCs from the AITD patients experienced a partial transition to a mesenchymal phenotype, preserving epithelial characteristics associated with a disruption in PC, which might contribute to AITD pathogenesis.
Collapse
Affiliation(s)
- Pablo Sacristán-Gómez
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
| | - Ana Serrano-Somavilla
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
| | - Lía Castro-Espadas
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
| | - Nuria Sánchez de la Blanca Carrero
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
| | - Miguel Sampedro-Núñez
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
| | - José Luis Muñoz-De-Nova
- Department of General and Digestive Surgery, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
| | - Francisca Molina-Jiménez
- Gastroenterology Research Unit, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
| | - Alejandra Rosell
- Pathology Unit, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
| | - Mónica Marazuela
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
| | - Rebeca Martínez-Hernández
- Department of Endocrinology, Hospital Universitario de la Princesa, Instituto de Investigación Princesa, Universidad Autónoma de Madrid, C/Diego de León 62, 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER GCV14/ER/12), 28029 Madrid, Spain
- Faculty of Medicine, Universidad San Pablo CEU, Urbanización Montepríncipe, Alcorcón, 28925 Madrid, Spain
| |
Collapse
|
13
|
Kurzawa‐Akanbi M, Whitfield P, Burté F, Bertelli PM, Pathak V, Doherty M, Hilgen B, Gliaudelytė L, Platt M, Queen R, Coxhead J, Porter A, Öberg M, Fabrikova D, Davey T, Beh CS, Georgiou M, Collin J, Boczonadi V, Härtlova A, Taggart M, Al‐Aama J, Korolchuk VI, Morris CM, Guduric‐Fuchs J, Steel DH, Medina RJ, Armstrong L, Lako M. Retinal pigment epithelium extracellular vesicles are potent inducers of age-related macular degeneration disease phenotype in the outer retina. J Extracell Vesicles 2022; 11:e12295. [PMID: 36544284 PMCID: PMC9772497 DOI: 10.1002/jev2.12295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 11/18/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness. Vision loss is caused by the retinal pigment epithelium (RPE) and photoreceptors atrophy and/or retinal and choroidal angiogenesis. Here we use AMD patient-specific RPE cells with the Complement Factor H Y402H high-risk polymorphism to perform a comprehensive analysis of extracellular vesicles (EVs), their cargo and role in disease pathology. We show that AMD RPE is characterised by enhanced polarised EV secretion. Multi-omics analyses demonstrate that AMD RPE EVs carry RNA, proteins and lipids, which mediate key AMD features including oxidative stress, cytoskeletal dysfunction, angiogenesis and drusen accumulation. Moreover, AMD RPE EVs induce amyloid fibril formation, revealing their role in drusen formation. We demonstrate that exposure of control RPE to AMD RPE apical EVs leads to the acquisition of AMD features such as stress vacuoles, cytoskeletal destabilization and abnormalities in the morphology of the nucleus. Retinal organoid treatment with apical AMD RPE EVs leads to disrupted neuroepithelium and the appearance of cytoprotective alpha B crystallin immunopositive cells, with some co-expressing retinal progenitor cell markers Pax6/Vsx2, suggesting injury-induced regenerative pathways activation. These findings indicate that AMD RPE EVs are potent inducers of AMD phenotype in the neighbouring RPE and retinal cells.
Collapse
Affiliation(s)
- Marzena Kurzawa‐Akanbi
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Phillip Whitfield
- Glasgow Polyomics and Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Florence Burté
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Pietro Maria Bertelli
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Varun Pathak
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Mary Doherty
- Lipidomics Research FacilityUniversity of the Highlands and IslandsInvernessUK
| | - Birthe Hilgen
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Lina Gliaudelytė
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | - Rachel Queen
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Jonathan Coxhead
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Andrew Porter
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Maria Öberg
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Daniela Fabrikova
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
| | - Tracey Davey
- Electron Microscopy Research ServicesNewcastle UniversityNewcastle upon TyneUK
| | - Chia Shyan Beh
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Maria Georgiou
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Joseph Collin
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Veronika Boczonadi
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Anetta Härtlova
- Institute of Biomedicine, Department of Microbiology and Immunology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- The Institute of Medical Microbiology and HygieneUniversity Medical Center Freiburg (Universitätklinikum Freiburg)FreiburgGermany
| | - Michael Taggart
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Jumana Al‐Aama
- Faculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Christopher M Morris
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Jasenka Guduric‐Fuchs
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - David H Steel
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Reinhold J Medina
- The Welcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Lyle Armstrong
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
14
|
Extracellular vimentin mimics VEGF and is a target for anti-angiogenic immunotherapy. Nat Commun 2022; 13:2842. [PMID: 35606362 PMCID: PMC9126915 DOI: 10.1038/s41467-022-30063-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Collapse
|
15
|
Zhang X, Zhang Q, Shan Y, Xiao J, Cheng J, Ye F, Sai Y. Transcriptomic investigation of the effects of TDCPP on PC12 and GC2 cells with experimental validation. Gene X 2022; 822:146349. [PMID: 35182677 DOI: 10.1016/j.gene.2022.146349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 11/25/2022] Open
Abstract
TDCPP is a flame retardant which has nervous and reproductive toxicity. Although there is a close association between nervous and reproductive system, the exact toxic mechanism of TDCPP in these systems is still seldom, especially in a genome scale. In this study, we explored the transcriptomic landscape of TDCPP in PC12 and GC2 cells using RNAseq method. A total of 465 co-differential expressed genes were found. These genes were mainly enriched in extra-cellular matrix, cell adhesion, cell cycle arrest, oxidoreductase activity GO terms, and PI3K/AKT, focal adhesion, ECM-receptor interaction KEGG pathways. Hub genes (ANXA1, COL27A1, GAS6, GNB4 and THBS1) were extracted using STRING and confirmed by qPCR experiment. Vimentin, HSPA5 and Caspase3 were proved to be responsible to TDCPP in GC2 and PC12 cells. Knockdown assay in PC12 cells showed that these hub genes could also affect the protein expression of vimentin, HSPA5 and Caspase3. In summary, TDCPP might exert its toxic effect through disturbing focal adhesion, ECM-receptor interaction and PI3K/Akt pathways. One of the mechanisms could be influence on the cytoskeleton (vimentin), ER stress (HSPA5) and apoptosis (Caspase3). The sequence data in this study might be a useful resource for future TDCPP related researches.
Collapse
Affiliation(s)
- Xi Zhang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qifu Zhang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yaohui Shan
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jingsong Xiao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jin Cheng
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Feng Ye
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Sai
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
16
|
Surolia R, Antony VB. Pathophysiological Role of Vimentin Intermediate Filaments in Lung Diseases. Front Cell Dev Biol 2022; 10:872759. [PMID: 35573702 PMCID: PMC9096236 DOI: 10.3389/fcell.2022.872759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Vimentin intermediate filaments, a type III intermediate filament, are among the most widely studied IFs and are found abundantly in mesenchymal cells. Vimentin intermediate filaments localize primarily in the cytoplasm but can also be found on the cell surface and extracellular space. The cytoplasmic vimentin is well-recognized for its role in providing mechanical strength and regulating cell migration, adhesion, and division. The post-translationally modified forms of Vimentin intermediate filaments have several implications in host-pathogen interactions, cancers, and non-malignant lung diseases. This review will analyze the role of vimentin beyond just the epithelial to mesenchymal transition (EMT) marker highlighting its role as a regulator of host-pathogen interactions and signaling pathways for the pathophysiology of various lung diseases. In addition, we will also examine the clinically relevant anti-vimentin compounds and antibodies that could potentially interfere with the pathogenic role of Vimentin intermediate filaments in lung disease.
Collapse
Affiliation(s)
| | - Veena B. Antony
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
17
|
Interplay between HTRA1 and classical signalling pathways in organogenesis and diseases. Saudi J Biol Sci 2022; 29:1919-1927. [PMID: 35531175 PMCID: PMC9072889 DOI: 10.1016/j.sjbs.2021.11.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/05/2021] [Accepted: 11/17/2021] [Indexed: 11/20/2022] Open
Abstract
The high temperature requirement factor A1 (HTRA1) is a serine protease which modulates an array of signalling pathways driving basal biological processes. HTRA1 plays a significant role in cell proliferation, migration and fate determination, in addition to controlling protein aggregates through refolding, translocation or degradation. The mutation of HTRA1 has been implicated in a plethora of disorders and this has also led to its growing interest as drug therapy target. This review details the involvement of HTRA1 in certain signalling pathways, namely the transforming growth factor beta (TGF-β), canonical Wingless/Integrated (WNT) and NOTCH signalling pathways during organogenesis and various disease pathogenesis such as preeclampsia, age-related macular degeneration (AMD), small vessel disease and cancer. We have also explored possible avenues of exploiting the serine proteases for therapeutic management of these disorders.
Collapse
|
18
|
Weghorst F, Mirzakhanyan Y, Hernandez KL, Gershon PD, Cramer KS. Non-Apoptotic Caspase Activity Preferentially Targets a Novel Consensus Sequence Associated With Cytoskeletal Proteins in the Developing Auditory Brainstem. Front Cell Dev Biol 2022; 10:844844. [PMID: 35330912 PMCID: PMC8940215 DOI: 10.3389/fcell.2022.844844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/28/2022] [Indexed: 11/24/2022] Open
Abstract
The auditory brainstem relies on precise circuitry to facilitate sound source localization. In the chick, the development of this specialized circuitry requires non-apoptotic activity of caspase-3, for which we previously identified several hundred proteolytic substrates. Here we tested whether the sequence of the caspase cleavage site differentially encodes proteolytic preference in apoptotic and non-apoptotic contexts. We constructed a consensus sequence for caspase activity in the non-apoptotic chick auditory brainstem comprising the four residues N-terminal to the cleavage site: IX(G/R)D↓ where X represents no significant enrichment and ↓ represents the cleavage site. We identified GO terms significantly enriched among caspase substrates containing motifs found in the above consensus sequence. (G/R)D↓ was associated with the term “Structural Constituent of Cytoskeleton” (SCoC), suggesting that SCoC proteins may be specifically targeted by caspase activity during non-apoptotic developmental processes. To ascertain whether this consensus sequence was specific to the non-apoptotic auditory brainstem at embryonic day (E) 10, we used protein mass spectrometry of brainstems harvested at a time when auditory brainstem neurons undergo apoptotic cell death (E13). The apoptotic motif VD was significantly enriched among E13 cleavage sites, indicating that motif preference at the P2 subsite had shifted toward the canonical caspase consensus sequence. Additionally, Monte Carlo simulations revealed that only the GD motif was associated with SCoC substrates in the apoptotic auditory brainstem, indicating that GD encodes specificity for SCoC proteins in both non-apoptotic and apoptotic contexts, despite not being preferred in the latter. Finally, to identify candidate human non-apoptotic consensus sequences, we used Monte Carlo analyses to determine motifs and motif pairs associated with SCoC caspase substrates in the Degrabase, a database of cleavage sites in human apoptotic cell lines. We found 11 motifs significantly associated with SCoC proteolysis, including IXXD and GD. We employed a stepwise method to select motif pairs that optimized SCoC specificity for a given coverage of SCoC cleavage events, yielding 11 motif pairs likely to be preferred in SCoC-directed human non-apoptotic caspase consensus sequences. GD + IXXD was among these motif pairs, suggesting a conservation of non-apoptotic consensus sites among vertebrates.
Collapse
Affiliation(s)
- Forrest Weghorst
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA, United States
| | - Yeva Mirzakhanyan
- Department of Molecular Biology and Biochemistry, UC Irvine, Irvine, CA, United States
| | | | - Paul D Gershon
- Department of Molecular Biology and Biochemistry, UC Irvine, Irvine, CA, United States
| | - Karina S Cramer
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA, United States
| |
Collapse
|
19
|
Hashemi Karoii D, Azizi H. A review of protein-protein interaction and signaling pathway of Vimentin in cell regulation, morphology and cell differentiation in normal cells. J Recept Signal Transduct Res 2022; 42:512-520. [PMID: 35296221 DOI: 10.1080/10799893.2022.2047199] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Vimentin intermediate filament (VIF) is an essential cytoskeleton component. It shows dynamically changing expression patterns throughout various phases of the differentiation process, suggesting that the protein is physiologically important. Vimentin's essential functions have recently been clear, so Vimentin-deficient of animals was described as a change of morphology and signaling pathway. Recent research has discovered many vital roles for Vimentin that were previously unknown. VIF emerges as an organizer of many essential proteins involved in movement and cell signaling. The highly dynamic and complicated phosphorylation of VIF seems to be a regulator mechanism for various activities. Changes in IF expression patterns are often linked with cancer progression, especially those leading to enhanced invasion and cellular migration. This review will discuss the function of Vimentin intermediate filaments in normal cell physiology, cell adhesion structures, cell shape, and signaling pathways. The genes interaction and gene network linked with Vimentin will be discussed in more studies. However, research aimed at understanding the function of Vimentin in different signaling cascades and gene interactions might offer novel methods for creating therapeutic medicines. Enrichr GEO datasets used gene ontology (GO) and pathway enrichment analyses. STRING online was used to predict the functional connections of proteins-proteins, followed by Cytoscape analysis to find the master genes. Cytoscape and STRING research revealed that eight genes, Fas, Casp8, Casp6, Fadd, Ripk1, Des, Tnnc2, and Tnnt3, were required for protein-protein interactions with Vimentin genes involved in cell differentiation.
Collapse
Affiliation(s)
- Danial Hashemi Karoii
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| |
Collapse
|
20
|
Epithelial–Fibroblast Crosstalk Protects against Acidosis-Induced Inflammatory and Fibrotic Alterations. Biomedicines 2022; 10:biomedicines10030681. [PMID: 35327483 PMCID: PMC8945333 DOI: 10.3390/biomedicines10030681] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/21/2022] Open
Abstract
Pathogenesis of chronic kidney disease (CKD) is accompanied by extracellular acidosis inflammation, fibrosis and epithelial-to-mesenchymal transition (EMT). The aim of this study was to assess the influence of acidosis on tubule epithelial cells (NRK-52E) and fibroblasts (NRK-49F) in dependence of cellular crosstalk. NRK-52E and NRK-49F were used in mono- and co-cultures, and were treated with acidic media (pH 6.0) for 48 h. The intracellular proteins were measured by Western blot. Secreted proteins were measured by ELISA. Distribution of E-cadherin was assessed by immunofluorescence and epithelial barrier function by FITC-dextran diffusion. Inflammation: Acidosis led to an increase in COX-2 in NRK-52E and TNF in NRK-49F in monoculture. In co-culture, this effect was reversed. EMT: Acidosis led to an increase in vimentin protein in both cell lines, whereas in co-culture, the effect was abolished. In NRK-52E, the E-cadherin expression was unchanged, but subcellular E-cadherin showed a disturbed distribution, and cellular barrier function was decreased. Fibrosis: Monoculture acidosis led to an increased secretion of collagen I and fibronectin in NRK-52E and collagen I in NRK-49F. In co-culture, the total collagen I secretion was unchanged, and fibronectin secretion was decreased. Intercellular crosstalk between epithelial cells and fibroblasts has a protective function regarding the development of acidosis-induced damage.
Collapse
|
21
|
Ostrowska-Podhorodecka Z, Ding I, Norouzi M, McCulloch CA. Impact of Vimentin on Regulation of Cell Signaling and Matrix Remodeling. Front Cell Dev Biol 2022; 10:869069. [PMID: 35359446 PMCID: PMC8961691 DOI: 10.3389/fcell.2022.869069] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Vimentin expression contributes to cellular mechanoprotection and is a widely recognized marker of fibroblasts and of epithelial-mesenchymal transition. But it is not understood how vimentin affects signaling that controls cell migration and extracellular matrix (ECM) remodeling. Recent data indicate that vimentin controls collagen deposition and ECM structure by regulating contractile force application to the ECM and through post-transcriptional regulation of ECM related genes. Binding of cells to the ECM promotes the association of vimentin with cytoplasmic domains of adhesion receptors such as integrins. After initial adhesion, cell-generated, myosin-dependent forces and signals that impact vimentin structure can affect cell migration. Post-translational modifications of vimentin determine its adaptor functions, including binding to cell adhesion proteins like paxillin and talin. Accordingly, vimentin regulates the growth, maturation and adhesive strength of integrin-dependent adhesions, which enables cells to tune their attachment to collagen, regulate the formation of cell extensions and control cell migration through connective tissues. Thus, vimentin tunes signaling cascades that regulate cell migration and ECM remodeling. Here we consider how specific properties of vimentin serve to control cell attachment to the underlying ECM and to regulate mesenchymal cell migration and remodeling of the ECM by resident fibroblasts.
Collapse
|
22
|
Vimentin: Regulation and pathogenesis. Biochimie 2022; 197:96-112. [DOI: 10.1016/j.biochi.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
|
23
|
Abstract
Apoptosis plays a key role in removing abnormal or senescent cells, maintaining the overall health of the tissue, and coordinating individual development. Recently, it has been discovered that the intracellular cytoskeleton plays a role in the apoptotic process. In addition, the regulatory role of extracellular matrix (ECM) fibrous proteins, which can be considered as the extracellular skeleton, in the process of apoptosis is rarely summarized. In this review, we collect the latest knowledge about how fibrous proteins inside and outside cells regulate apoptosis. We describe how ECM fibrous proteins participate in the regulation of death receptor and mitochondrial pathways through various signaling cascades mediated by integrins. We then explore the molecular mechanisms by which intracellular intermediate filaments regulate cell apoptosis by regulating death receptors on the cell membrane surface. Similarly, we report on novel supporting functions of microtubules in the execution phase of apoptosis and discuss their formation mechanisms. Finally, we discuss that the polypeptide fragments formed by caspase degradation of ECM fibrous proteins and intracellular intermediate filament act as local regulatory signals to play different regulatory roles in apoptosis.
Collapse
Affiliation(s)
- Jia-Hao Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Mogre S, Makani V, Pradhan S, Devre P, More S, Vaidya M, Dmello C. Biomarker Potential of Vimentin in Oral Cancers. Life (Basel) 2022; 12:150. [PMID: 35207438 PMCID: PMC8879320 DOI: 10.3390/life12020150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 12/31/2022] Open
Abstract
Oral carcinogenesis is a multistep process. As much as 5% to 85% of oral tumors can develop from potentially malignant disorders (PMD). Although the oral cavity is accessible for visual examination, the ability of current clinical or histological methods to predict the lesions that can progress to malignancy is limited. Thus, developing biological markers that will serve as an adjunct to histodiagnosis has become essential. Our previous studies comprehensively demonstrated that aberrant vimentin expression in oral premalignant lesions correlates to the degree of malignancy. Likewise, overwhelming research from various groups show a substantial contribution of vimentin in oral cancer progression. In this review, we have described studies on vimentin in oral cancers, to make a compelling case for vimentin as a prognostic biomarker.
Collapse
Affiliation(s)
- Saie Mogre
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Vidhi Makani
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India; (V.M.); (S.P.); (P.D.)
| | - Swapnita Pradhan
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India; (V.M.); (S.P.); (P.D.)
| | - Pallavi Devre
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India; (V.M.); (S.P.); (P.D.)
| | - Shyam More
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Milind Vaidya
- Vaidya Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India; (V.M.); (S.P.); (P.D.)
| | - Crismita Dmello
- Department of Neurological Surgery, Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
25
|
van Asperen JV, Robe PA, Hol EM. GFAP Alternative Splicing and the Relevance for Disease – A Focus on Diffuse Gliomas. ASN Neuro 2022; 14:17590914221102065. [PMID: 35673702 PMCID: PMC9185002 DOI: 10.1177/17590914221102065] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament protein that is
characteristic for astrocytes and neural stem cells, and their malignant analogues in
glioma. Since the discovery of the protein 50 years ago, multiple alternative splice
variants of the GFAP gene have been discovered, leading to different GFAP isoforms. In
this review, we will describe GFAP isoform expression from gene to protein to network,
taking the canonical isoforms GFAPα and the main alternative variant GFAPδ as the starting
point. We will discuss the relevance of studying GFAP and its isoforms in disease, with a
specific focus on diffuse gliomas.
Collapse
Affiliation(s)
- Jessy V. van Asperen
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Pierre A.J.T. Robe
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, University Utrecht, Utrecht, The Netherlands
| | - Elly M. Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
26
|
Usman S, Waseem NH, Nguyen TKN, Mohsin S, Jamal A, Teh MT, Waseem A. Vimentin Is at the Heart of Epithelial Mesenchymal Transition (EMT) Mediated Metastasis. Cancers (Basel) 2021; 13:4985. [PMID: 34638469 PMCID: PMC8507690 DOI: 10.3390/cancers13194985] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible plethora of molecular events where epithelial cells gain the phenotype of mesenchymal cells to invade the surrounding tissues. EMT is a physiological event during embryogenesis (type I) but also happens during fibrosis (type II) and cancer metastasis (type III). It is a multifaceted phenomenon governed by the activation of genes associated with cell migration, extracellular matrix degradation, DNA repair, and angiogenesis. The cancer cells employ EMT to acquire the ability to migrate, resist therapeutic agents and escape immunity. One of the key biomarkers of EMT is vimentin, a type III intermediate filament that is normally expressed in mesenchymal cells but is upregulated during cancer metastasis. This review highlights the pivotal role of vimentin in the key events during EMT and explains its role as a downstream as well as an upstream regulator in this highly complex process. This review also highlights the areas that require further research in exploring the role of vimentin in EMT. As a cytoskeletal protein, vimentin filaments support mechanical integrity of the migratory machinery, generation of directional force, focal adhesion modulation and extracellular attachment. As a viscoelastic scaffold, it gives stress-bearing ability and flexible support to the cell and its organelles. However, during EMT it modulates genes for EMT inducers such as Snail, Slug, Twist and ZEB1/2, as well as the key epigenetic factors. In addition, it suppresses cellular differentiation and upregulates their pluripotent potential by inducing genes associated with self-renewability, thus increasing the stemness of cancer stem cells, facilitating the tumour spread and making them more resistant to treatments. Several missense and frameshift mutations reported in vimentin in human cancers may also contribute towards the metastatic spread. Therefore, we propose that vimentin should be a therapeutic target using molecular technologies that will curb cancer growth and spread with reduced mortality and morbidity.
Collapse
Affiliation(s)
- Saima Usman
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Naushin H. Waseem
- UCL Institute of Ophthalmology, 11-43 Bath Str., London EC1V 9EL, UK;
| | - Thuan Khanh Ngoc Nguyen
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Sahar Mohsin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Ahmad Jamal
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Muy-Teck Teh
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Ahmad Waseem
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| |
Collapse
|
27
|
Tripathi D, Kulkarni S. Butein induces intrinsic pathway of apoptosis, vimentin proteolysis, and inhibition of cancer stem cell population in a human papillary thyroid cancer cell line. Toxicol In Vitro 2021; 77:105244. [PMID: 34481015 DOI: 10.1016/j.tiv.2021.105244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/16/2023]
Abstract
Epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) play an essential role in metastasis of papillary thyroid cancer (PTC). Further mesenchymal marker vimentin is linked with metastasis and cancer stem cell generation. Hence, inhibition of EMT and effective elimination of CSCs offers a novel target for the development of new therapeutic agents. The present study observed that at lower concentration, butein, a major bioactive chalcone, significantly inhibits NPA (papillary thyroid cancer cell line) cell migration and reduces extracellular acidification rate (ECAR) an indicator of enhanced glycolysis, required for cell migration. Additionally, at lower concentrations, butein treatment also suppresses vimentin phosphorylation, an essential step in cell migration, proving its potential against cell migration. Phosphorylation of vimentin is crucial in the protection of vimentin from caspase-mediated proteolysis. Interestingly, butein activates caspase-3 for the apoptosis execution at higher concentration; hence, total levels of vimentin were investigated. Butein induces caspase-3 mediated proteolysis of vimentin. Vimentin and glycolysis are essential for maintaining CSCs; therefore, aldeflour assay and side population assay were performed to investigate the effect of butein on CSCs. Our data suggest butein mediates the reduction in CSCs population. Here we report a novel mechanism of butein mediated inhibition of NPA cells migration by suppressing vimentin phosphorylation and its subsequent proteolysis. Collectively our data suggest the potential of butein as an innovative anticancer therapeutic agent for PTC management.
Collapse
Affiliation(s)
- Devavrat Tripathi
- Radiation Medicine Centre, Bhabha Atomic Research Centre, c/o TMH Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India
| | - Savita Kulkarni
- Radiation Medicine Centre, Bhabha Atomic Research Centre, c/o TMH Annexe, Parel, Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, India.
| |
Collapse
|
28
|
Muñoz EN, Rivera HM, Gómez LA. Changes in cytoarchitecture and mobility in B16F1 melanoma cells induced by 5-Br-2'-dU coincide with Rock2, miRNAs 138-5p and 455-3p reciprocal expressions. Biochem Biophys Rep 2021; 27:101027. [PMID: 34159262 PMCID: PMC8202345 DOI: 10.1016/j.bbrep.2021.101027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 11/23/2022] Open
Abstract
ROCK2 is a protein involved in the restructuring of the cytoskeleton in cell adhesion and contractibility processes. miR-138-5p and miR-455-3p regulate Rock2 expression, cell proliferation, migration, and invasion in different experimental cell models. However, their participation in the cytoarchitecture and mobility of B16F1 melanoma cells exposed to 5-Br-2'-dU is partially known. This work aimed to analyze ROCK2 and miRs 138-5p and 455-3p expression associated with morphological and mobility changes of B16F1 mouse melanoma cells exposed to the thymidine analog 5-Bromo-2'-deoxyuridine (5-Br-2'-dU). We observed an increase (2.2X n = 3, p < 0.05) in the cell area, coinciding with an increase in cell diameter (1.27X n = 3, p < 0.05), as well as greater cell granularity, capacity for circularization, adhesion, which was associated with more significant polymerization of F-actin, collapsed in the intermediate filaments of vimentin (VIM), and coinciding with a decrease in migration (87%). Changes coincided with a decrease in Rock2 mRNA expression (2.88X n = 3, p < 0.05), increased vimentin and a reciprocal decrease in miR-138-5p (1.8X), and an increase in miR-455-3p (2.39X). The Rock2 kinase inhibitor Y27632 partially rescued these changes. These results suggest ROCK2 and VIM regulate the morphological and mobility changes of B16 melanoma cells after exposure to 5-Br-2'-dU, and its expression may be reciprocally regulated, at least in part, by miR-138-5p and miR-455-3p.
Collapse
Affiliation(s)
- Esther Natalia Muñoz
- Molecular Physiology Group, Scientific and Technological Research, Public Health Research, Instituto Nacional de Salud de Colombia, Bogotá, D.C., Colombia
- Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, D.C., Colombia
| | - Hernán Mauricio Rivera
- Molecular Physiology Group, Scientific and Technological Research, Public Health Research, Instituto Nacional de Salud de Colombia, Bogotá, D.C., Colombia
| | - Luis Alberto Gómez
- Molecular Physiology Group, Scientific and Technological Research, Public Health Research, Instituto Nacional de Salud de Colombia, Bogotá, D.C., Colombia
- Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, D.C., Colombia
| |
Collapse
|
29
|
Rosner M, Hengstschläger M. Three-dimensional migration of human amniotic fluid stem cells involves mesenchymal and amoeboid modes and is regulated by mTORC1. STEM CELLS (DAYTON, OHIO) 2021; 39:1718-1732. [PMID: 34331786 PMCID: PMC9291078 DOI: 10.1002/stem.3441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022]
Abstract
Three‐dimensional (3D) cell migration is an integral part of many physiologic processes. Although being well studied in the context of adult tissue homeostasis and cancer development, remarkably little is known about the invasive behavior of human stem cells. Using two different kinds of invasion assays, this study aimed at investigating and characterizing the 3D migratory capacity of human amniotic fluid stem cells (hAFSCs), a well‐established fetal stem cell type. Eight hAFSC lines were found to harbor pronounced potential to penetrate basement membrane (BM)‐like matrices. Morphological examination and inhibitor approaches revealed that 3D migration of hAFSCs involves both the matrix metalloprotease‐dependent mesenchymal, elongated mode and the Rho‐associated protein kinase‐dependent amoeboid, round mode. Moreover, hAFSCs could be shown to harbor transendothelial migration capacity and to exhibit a motility‐associated marker expression pattern. Finally, the potential to cross extracellular matrix was found to be induced by mTORC1‐activating growth factors and reduced by blocking mTORC1 activity. Taken together, this report provides the first demonstration that human stem cells exhibit mTORC1‐dependent invasive capacity and can concurrently make use of mesenchymal and amoeboid 3D cell migration modes, which represents an important step toward the full biological characterization of fetal human stem cells with relevance to both developmental research and stem cell‐based therapy.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Evaluation of the effects of phosphorylation of synthetic peptide substrates on their cleavage by caspase-3 and -7. Biochem J 2021; 478:2233-2245. [PMID: 34037204 DOI: 10.1042/bcj20210255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/02/2023]
Abstract
Caspases are a family of enzymes that play roles in cell death and inflammation. It has been suggested that in the execution phase of the apoptotic pathway, caspase-3, -6 and -7 are involved. The substrate specificities of two proteases (caspases 3 and 7) are highly similar, which complicates the design of compounds that selectively interact with a single enzyme exclusively. The recognition of residues other than Asp in the P1 position of the substrate by caspase-3/-7 has been reported, promoting interest in the effects of phosphorylation of amino acids in the direct vicinity of the scissile bond. To evaluate conflicting reports on this subject, we synthesized a series of known caspase-3 and -7 substrates and phosphorylated analogs, performed enzyme kinetic assays and mapped the peptide cleavage sites using internally quenched fluorescent peptide substrates. Caspases 3 and 7 will tolerate pSer at the P1 position but only poorly at the P2' position. Our investigation demonstrates the importance of peptide length and composition in interpreting sequence/activity relationships. Based on the results, we conclude that the relationship between caspase-3/-7 and their substrates containing phosphorylated amino acids might depend on the steric conditions and not be directly connected with ionic interactions. Thus, the precise effect of phospho-amino acid residues located in the vicinity of the cleaved bond on the regulation of the substrate specificity of caspases remains difficult to predict. Our observations allow to predict that natural phosphorylated proteins may be cleaved by caspases, but only when extended substrate binding site interactions are satisfied.
Collapse
|
31
|
Rare CASP6N73T variant associated with hippocampal volume exhibits decreased proteolytic activity, synaptic transmission defect, and neurodegeneration. Sci Rep 2021; 11:12695. [PMID: 34135352 PMCID: PMC8209045 DOI: 10.1038/s41598-021-91367-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/25/2021] [Indexed: 01/22/2023] Open
Abstract
Caspase-6 (Casp6) is implicated in Alzheimer disease (AD) cognitive impairment and pathology. Hippocampal atrophy is associated with cognitive impairment in AD. Here, a rare functional exonic missense CASP6 single nucleotide polymorphism (SNP), causing the substitution of asparagine with threonine at amino acid 73 in Casp6 (Casp6N73T), was associated with hippocampal subfield CA1 volume preservation. Compared to wild type Casp6 (Casp6WT), recombinant Casp6N73T altered Casp6 proteolysis of natural substrates Lamin A/C and α-Tubulin, but did not alter cleavage of the Ac-VEID-AFC Casp6 peptide substrate. Casp6N73T-transfected HEK293T cells showed elevated Casp6 mRNA levels similar to Casp6WT-transfected cells, but, in contrast to Casp6WT, did not accumulate active Casp6 subunits nor show increased Casp6 enzymatic activity. Electrophysiological and morphological assessments showed that Casp6N73T recombinant protein caused less neurofunctional damage and neurodegeneration in hippocampal CA1 pyramidal neurons than Casp6WT. Lastly, CASP6 mRNA levels were increased in several AD brain regions confirming the implication of Casp6 in AD. These studies suggest that the rare Casp6N73T variant may protect against hippocampal atrophy due to its altered catalysis of natural protein substrates and intracellular instability thus leading to less Casp6-mediated damage to neuronal structure and function.
Collapse
|
32
|
Pla I, Sanchez A, Pors SE, Pawlowski K, Appelqvist R, Sahlin KB, Poulsen LLC, Marko-Varga G, Andersen CY, Malm J. Proteome of fluid from human ovarian small antral follicles reveals insights in folliculogenesis and oocyte maturation. Hum Reprod 2021; 36:756-770. [PMID: 33313811 PMCID: PMC7891813 DOI: 10.1093/humrep/deaa335] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION Is it possible to identify by mass spectrometry a wider range of proteins and key proteins involved in folliculogenesis and oocyte growth and development by studying follicular fluid (FF) from human small antral follicles (hSAF)? SUMMARY ANSWER The largest number of proteins currently reported in human FF was identified in this study analysing hSAF where several proteins showed a strong relationship with follicular developmental processes. WHAT IS KNOWN ALREADY Protein composition of human ovarian FF constitutes the microenvironment for oocyte development. Previous proteomics studies have analysed fluids from pre-ovulatory follicles, where large numbers of plasma constituents are transferred through the follicular basal membrane. This attenuates the detection of low abundant proteins, however, the basal membrane of small antral follicles is less permeable, making it possible to detect a large number of proteins, and thereby offering further insights in folliculogenesis. STUDY DESIGN, SIZE, DURATION Proteins in FF from unstimulated hSAF (size 6.1 ± 0.4 mm) were characterised by mass spectrometry, supported by high-throughput and targeted proteomics and bioinformatics. The FF protein profiles from hSAF containing oocytes, capable or not of maturing to metaphase II of the second meiotic division during an IVM (n = 13, from 6 women), were also analysed. PARTICIPANTS/MATERIALS, SETTING, METHODS We collected FF from hSAF of ovaries that had been surgically removed from 31 women (∼28.5 years old) undergoing unilateral ovariectomy for fertility preservation. MAIN RESULTS AND THE ROLE OF CHANCE In total, 2461 proteins were identified, of which 1108 identified for the first time in FF. Of the identified proteins, 24 were related to follicular regulatory processes. A total of 35 and 65 proteins were down- and up-regulated, respectively, in fluid from hSAF surrounding oocytes capable of maturing (to MII). We found that changes at the protein level occur already in FF from small antral follicles related to subsequent oocyte maturation. LIMITATIONS, REASONS FOR CAUTION A possible limitation of our study is the uncertainty of the proportion of the sampled follicles that are undergoing atresia. Although the FF samples were carefully aspirated and processed to remove possible contaminants, we cannot ensure the absence of some proteins derived from cellular lysis provoked by technical reasons. WIDER IMPLICATIONS OF THE FINDINGS This study is, to our knowledge, the first proteomics characterisation of FF from hSAF obtained from women in their natural menstrual cycle. We demonstrated that the analysis by mass spectrometry of FF from hSAF allows the identification of a greater number of proteins compared to the results obtained from previous analyses of larger follicles. Significant differences found at the protein level in hSAF fluid could predict the ability of the enclosed oocyte to sustain meiotic resumption. If this can be confirmed in further studies, it demonstrates that the viability of the oocyte is determined early on in follicular development and this may open up new pathways for augmenting or attenuating subsequent oocyte viability in the pre-ovulatory follicle ready to undergo ovulation. STUDY FUNDING/COMPETING INTEREST(S) The authors thank the financial support from ReproUnion, which is funded by the Interreg V EU programme. No conflict of interest was reported by the authors. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Indira Pla
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, 2100 Copenhagen, Denmark
| | - Krzysztof Pawlowski
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden.,Department of Experimental Design and Bioinformatics, Faculty of Agriculture and Biology, Warsaw University of Life Sciences SGGW, Warszawa 02-787, Poland
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - K Barbara Sahlin
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| | - Liv La Cour Poulsen
- Fertility Clinic, Department of Gynaecology and Obstetrics, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - György Marko-Varga
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden.,First Department of Surgery, Tokyo Medical University, Shinjiku-ku, Japan
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, 2100 Copenhagen, Denmark
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 205 02 Malmö, Sweden.,Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, BMC D13, 221 84 Lund, Sweden
| |
Collapse
|
33
|
Prostaglandin F2 Alpha Triggers the Disruption of Cell Adhesion with Cytokeratin and Vimentin in Bovine Luteal Theca Cells. Animals (Basel) 2021; 11:ani11041073. [PMID: 33918916 PMCID: PMC8069824 DOI: 10.3390/ani11041073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 03/25/2021] [Indexed: 11/26/2022] Open
Abstract
Simple Summary Luteolysis is an important event in the control of the corpus luteum function in bovines. However, some aspects of the luteolytic mechanism remain unclear. We evaluated changes in cell adhesion in luteal cells during regression of corpus luteum. Bovine luteal theca cells (LTCs) were treated in vitro with Prostaglandin F2 alpha (PGF2α). Cytokeratin, vimentin and desmoplakin proteins in LTCs were disrupted by PGF2α, affecting cell adhesion. These results suggest that PGF2α plays an important function in cell adhesion during the regression of corpus luteum. Abstract Intermediate filaments (IFs) maintain cell–cell adhesions and are involved in diverse cellular processes such as cytokinesis, cell migration and the maintenance of cell structure. In this study, we investigated the influence of prostaglandin F2 alpha (PGF2α) on cytokeratin and vimentin IFs, Rho-associated protein kinase (ROCK), and cell-cell adhesion in bovine luteal theca cells (LTCs). The luteal cells were isolated from bovine corpus luteum (CL), and the LTCs were treated with 0, 0.01, 0.1 and 1.0 mM PGF2α. Cytokeratin, vimentin and desmoplakin proteins were disrupted and the ROCK protein was significantly increased in PGF2α-treated LTCs. In addition, cell–cell adhesion was significantly (p < 0.05) decreased in the PGF2α-induced LTCs compared to control group (0 mM PGF2α). In conclusion, PGF2α affected the adhesion of cell to cell via disruption of desmoplakin, cytokeratin and vimentin, additionally increasing ROCK in bovine LTCs. These results may provide a better understanding of the mechanism of bovine CL regression.
Collapse
|
34
|
Xiao J, Chen L, Melander O, Orho-Melander M, Nilsson J, Borné Y, Engström G. Circulating Vimentin Is Associated With Future Incidence of Stroke in a Population-Based Cohort Study. Stroke 2021; 52:937-944. [PMID: 33535783 DOI: 10.1161/strokeaha.120.032111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE VIM (vimentin) is a cytoskeletal intermediate filament protein, which has been linked to atherosclerosis and thrombosis; both are important causes of stroke. We examined the relationship between circulating VIM and incidence of stroke, and if carotid plaque could modify the association in a prospective population-based cohort. METHODS This prospective study was based on the Malmö Diet and Cancer Cohort. A total of 4688 participants (39.7% men; mean age, 57.6 years) were examined and blood samples were collected between 1991 and 1994. Incidence of stroke was followed up to 2018. Cox' proportional hazards regression was used to assess the relationship between VIM and stroke. RESULTS During a mean follow-up of 22.0 years, a total of 528 subjects were diagnosed with stroke, among which 434 were ischemic stroke. Participants in the highest quartile (vs 1st quartile) had 1.34× higher risk of total stroke (95% CI, 1.03-1.74) and 1.47× higher of ischemic stroke (95% CI, 1.10-1.98) after adjustment for potential confounders. A significant interaction was found between carotid plaque and VIM with respect to incidence of both total stroke and ischemic stroke (P=0.041 and 0.011, respectively). After stratifying by carotid plaque, high VIM had stronger association with stroke in participants with carotid plaque, especially for the risk of ischemic stroke (adjusted hazard ratio,1.66 [95% CI, 1.23-2.25] for quartile 4 versus quartile 1 to 3). CONCLUSIONS VIM is positively associated with the incidence of stroke, especially in individuals with carotid plaque. Further studies are needed to confirm the observed associations.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China (J.X., L.C.).,Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China (J.X., L.C.)
| | - Olle Melander
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| | - Marju Orho-Melander
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| | - Jan Nilsson
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| | - Yan Borné
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden (J.X., O.M., M.O.-M., J.N., Y.B., G.E.)
| |
Collapse
|
35
|
Abdel-Wahab BA, Salem SY, Mohammed HM, Mohammed NA, Hetta HF. The role of vimentin, Connexin-43 proteins, and oxidative stress in the protective effect of propranolol against clozapine-induced myocarditis and apoptosis in rats. Eur J Pharmacol 2021; 890:173645. [PMID: 33098837 DOI: 10.1016/j.ejphar.2020.173645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/02/2020] [Accepted: 10/08/2020] [Indexed: 12/31/2022]
Abstract
Clozapine (CLZ) represents an effective treatment for resistant schizophrenia. However, myocarditis, recently reported in about 66% of the psychiatric patients treated with CLZ, has raised concerns about its safety. β-blocking agents have shown to be helpful in the management of myocarditis. Moreover, Vimentin (VIM) and Connexin-43 (CX43) are important structural proteins play key roles in cytoskeletal functions and cellular communication and have complex implications in pathophysiology. The present work aimed to study the mechanisms behind the protective effect of propranolol (PRO) against CLZ-induced myocarditis and the possible involvement of VIM and CX43. The effect of PRO (5 and 10 mg/kg, oral) on the myocarditis induced by CLZ (25 mg/kg/d, i. p.) treatment for 21 days in rats, was assessed biochemically, and immunohistochemically. CLZ treatment increased the serum levels of cardiac injury (CK-MP, LDH and cTn-I) and cardiac levels of oxidative stress (TBARS and NO) markers, proinflammatory cytokines (IL-1β and TNF-α), and mRNA expression of VIM and CX43 with decreased the antioxidant defenses (GSH and GSH-Px). Immunohistochemical study showed increased cardiac expression of VIM, CX43 and caspase-3 proteins. Coadministration of PRO with CLZ, dose-dependently decreased the biochemical and immunohistochemical hallmarks of CLZ-induced myocardial injury and significantly decreased mRNA expression of VIM and CX43. Taken together, our results demonstrate that the cardioprotective effects of PRO on CLZ-induced myocarditis are related in addition to its β-blocking activity to protection of myocardial VIM and CX43 proteins through antagonizing the CLZ-induced oxidative stress and inflammatory response, and preventing cell apoptosis.
Collapse
Affiliation(s)
- Basel A Abdel-Wahab
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Safaa Yousef Salem
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Hala Mostafa Mohammed
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
36
|
Patteson AE, Carroll RJ, Iwamoto DV, Janmey PA. The vimentin cytoskeleton: when polymer physics meets cell biology. Phys Biol 2020; 18:011001. [PMID: 32992303 PMCID: PMC8240483 DOI: 10.1088/1478-3975/abbcc2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The proper functions of tissues depend on the ability of cells to withstand stress and maintain shape. Central to this process is the cytoskeleton, comprised of three polymeric networks: F-actin, microtubules, and intermediate filaments (IFs). IF proteins are among the most abundant cytoskeletal proteins in cells; yet they remain some of the least understood. Their structure and function deviate from those of their cytoskeletal partners, F-actin and microtubules. IF networks show a unique combination of extensibility, flexibility and toughness that confers mechanical resilience to the cell. Vimentin is an IF protein expressed in mesenchymal cells. This review highlights exciting new results on the physical biology of vimentin intermediate filaments and their role in allowing whole cells and tissues to cope with stress.
Collapse
Affiliation(s)
- Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Robert J Carroll
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Daniel V Iwamoto
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Tomihara H, Carbone F, Perelli L, Huang JK, Soeung M, Rose JL, Robinson FS, Lissanu Deribe Y, Feng N, Takeda M, Inoue A, Poggetto ED, Deem AK, Maitra A, Msaouel P, Tannir NM, Draetta GF, Viale A, Heffernan TP, Bristow CA, Carugo A, Genovese G. Loss of ARID1A Promotes Epithelial-Mesenchymal Transition and Sensitizes Pancreatic Tumors to Proteotoxic Stress. Cancer Res 2020; 81:332-343. [PMID: 33158812 DOI: 10.1158/0008-5472.can-19-3922] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/19/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022]
Abstract
Cellular dedifferentiation is a key mechanism driving cancer progression. Acquisition of mesenchymal features has been associated with drug resistance, poor prognosis, and disease relapse in many tumor types. Therefore, successful targeting of tumors harboring these characteristics is a priority in oncology practice. The SWItch/Sucrose non-fermentable (SWI/SNF) chromatin remodeling complex has also emerged as a critical player in tumor progression, leading to the identification of several SWI/SNF complex genes as potential disease biomarkers and targets of anticancer therapies. AT-rich interaction domain-containing protein 1A (ARID1A) is a component of SWI/SNF, and mutations in ARID1A represent one of the most frequent molecular alterations in human cancers. ARID1A mutations occur in approximately 10% of pancreatic ductal adenocarcinomas (PDAC), but whether these mutations confer a therapeutic opportunity remains unclear. Here, we demonstrate that loss of ARID1A promotes an epithelial-mesenchymal transition (EMT) phenotype and sensitizes PDAC cells to a clinical inhibitor of HSP90, NVP-AUY922, both in vitro and in vivo. Although loss of ARID1A alone did not significantly affect proliferative potential or rate of apoptosis, ARID1A-deficient cells were sensitized to HSP90 inhibition, potentially by promoting the degradation of intermediate filaments driving EMT, resulting in cell death. Our results describe a mechanistic link between ARID1A defects and a quasi-mesenchymal phenotype, suggesting that deleterious mutations in ARID1A associated with protein loss exhibit potential as a biomarker for patients with PDAC who may benefit by HSP90-targeting drugs treatment. SIGNIFICANCE: This study identifies ARID1A loss as a promising biomarker for the identification of PDAC tumors that are potentially responsive to treatment with proteotoxic agents.
Collapse
Affiliation(s)
- Hideo Tomihara
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Federica Carbone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luigi Perelli
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Justin K Huang
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Houston, Houston, Texas
| | - Johnathon L Rose
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Houston, Houston, Texas
| | - Frederick S Robinson
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yonathan Lissanu Deribe
- Department of Thoracic and Cardio Surgery-Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ningping Feng
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mitsunobu Takeda
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Akira Inoue
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edoardo Del Poggetto
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angela K Deem
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Sheikh Ahmed Bin Zayed Al Nahyan Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy P Heffernan
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher A Bristow
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandro Carugo
- Therapeutics Discovery Division, TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Giannicola Genovese
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
38
|
Zheng K, Li Q, Lin D, Zong X, Luo X, Yang M, Yue X, Ma S. Peptidomic analysis of pilose antler and its inhibitory effect on triple-negative breast cancer at multiple sites. Food Funct 2020; 11:7481-7494. [PMID: 32789330 DOI: 10.1039/d0fo01531h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pilose antler (PA) is a traditional Chinese functional food that has been reported to inhibit breast cancer; however, the specific substances that exert this effect and the underlying mechanisms remain unknown. This study aims to identify the specific proteins in PA water-soluble polypeptides (PAWPs) that are involved in cancer inhibition and determine the effects of PAWPs on triple-negative breast cancer in mice. In this study, peptidomic analysis of 105 varieties of polypeptides from PAWPs was carried out using LC-MS, 22 of which had functions that could potentially suppress tumors, including endopeptidase inhibitors, metal ion-binding proteins, angiogenesis inhibitors, intercellular adhesion proteins, and extracellular matrix repair proteins. Furthermore, we showed that intragastric administration of PAWPs into mice inhibited the growth and metastasis of triple-negative 4T1 breast tumors. PAWPs activated the expression of cleaved-caspase3 and increased tumor apoptosis, resulting in the reduction of platelet-endothelial cell adhesion molecule (PECAM-1/CD31) expression and the number of blood vessels, as well as the inhibition of matrix metalloproteinase (MMP) 2 and 9, increasing the ratio of Cadherin-1 (CDH1)/Cadherin-2 (CDH2) and inhibiting epithelial-mesenchymal transition (EMT) in these tumors. Therefore, PAWPs inhibit the progression and metastasis of triple-negative 4T1 breast cancer at multiple key sites in mice and contain various tumor suppressor proteins that are potentially involved in these processes.
Collapse
Affiliation(s)
- Kexin Zheng
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Qilong Li
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Dongdong Lin
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Xiaoyan Zong
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| | - Xue Luo
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Mei Yang
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Xiqing Yue
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Shiliang Ma
- College of Food Science and Technology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China. and College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang, Liaoning 110866, China
| |
Collapse
|
39
|
Lo B, Marty-Gasset N, Pichereaux C, Bravo C, Manse H, Domitile R, Rémignon H. Proteomic Analysis of Two Weight Classes of Mule Duck " foie gras" at the End of an Overfeeding Period. Front Physiol 2020; 11:569329. [PMID: 33041868 PMCID: PMC7528769 DOI: 10.3389/fphys.2020.569329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/14/2020] [Indexed: 12/28/2022] Open
Abstract
The weight of the liver is one of the important selection criteria in the quality of “foie gras”. This factor is highly variable despite the fact that individuals are reared, overfed and slaughtered in the same way. In this study, we performed an analysis of the proteome profile of two weight classes of light (between 550 and 599 g) and heavy (more than 700 g) livers. For the analysis of the proteic extracts, a liquid chromatographic analysis coupled with mass spectrometry was carried out. In low-weight livers, aerobic energy metabolism, protein metabolism and lipid metabolism oriented toward export and beta-oxidation were overexpressed. On the contrary, high weight livers were characterized by anaerobic energy metabolism and a more active protein catabolism associated with cell apoptosis and reorganization of the cell structure.
Collapse
Affiliation(s)
- Bara Lo
- Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, Ecole Nationale Vétérinaire de Toulouse, Université de Toulouse, GENétique PHYsiologie et Systèmes d'Elevage, Castanet-Tolosan, France
| | - Nathalie Marty-Gasset
- Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, Ecole Nationale Vétérinaire de Toulouse, Université de Toulouse, GENétique PHYsiologie et Systèmes d'Elevage, Castanet-Tolosan, France
| | - Carole Pichereaux
- Centre National de la Recherche Scientifique, Fédération de Recherche (FR3450), Agrobiosciences, Interactions et Biodiversité, Toulouse, France.,Centre National de la Recherche Scientifique, Université de Toulouse - UPS, Institut de Pharmacologie et Biologie Structurale, Toulouse, France
| | - Céline Bravo
- Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, Ecole Nationale Vétérinaire de Toulouse, Université de Toulouse, GENétique PHYsiologie et Systèmes d'Elevage, Castanet-Tolosan, France
| | - Hélène Manse
- Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, Ecole Nationale Vétérinaire de Toulouse, Université de Toulouse, GENétique PHYsiologie et Systèmes d'Elevage, Castanet-Tolosan, France
| | | | - Hervé Rémignon
- Institut National de Recherche Pour l'Agriculture, l'Alimentation et l'Environnement, Ecole Nationale Vétérinaire de Toulouse, Université de Toulouse, GENétique PHYsiologie et Systèmes d'Elevage, Castanet-Tolosan, France
| |
Collapse
|
40
|
Ramos I, Stamatakis K, Oeste CL, Pérez-Sala D. Vimentin as a Multifaceted Player and Potential Therapeutic Target in Viral Infections. Int J Mol Sci 2020; 21:E4675. [PMID: 32630064 PMCID: PMC7370124 DOI: 10.3390/ijms21134675] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/17/2022] Open
Abstract
Vimentin is an intermediate filament protein that plays key roles in integration of cytoskeletal functions, and therefore in basic cellular processes such as cell division and migration. Consequently, vimentin has complex implications in pathophysiology. Vimentin is required for a proper immune response, but it can also act as an autoantigen in autoimmune diseases or as a damage signal. Although vimentin is a predominantly cytoplasmic protein, it can also appear at extracellular locations, either in a secreted form or at the surface of numerous cell types, often in relation to cell activation, inflammation, injury or senescence. Cell surface targeting of vimentin appears to associate with the occurrence of certain posttranslational modifications, such as phosphorylation and/or oxidative damage. At the cell surface, vimentin can act as a receptor for bacterial and viral pathogens. Indeed, vimentin has been shown to play important roles in virus attachment and entry of severe acute respiratory syndrome-related coronavirus (SARS-CoV), dengue and encephalitis viruses, among others. Moreover, the presence of vimentin in specific virus-targeted cells and its induction by proinflammatory cytokines and tissue damage contribute to its implication in viral infection. Here, we recapitulate some of the pathophysiological implications of vimentin, including the involvement of cell surface vimentin in interaction with pathogens, with a special focus on its role as a cellular receptor or co-receptor for viruses. In addition, we provide a perspective on approaches to target vimentin, including antibodies or chemical agents that could modulate these interactions to potentially interfere with viral pathogenesis, which could be useful when multi-target antiviral strategies are needed.
Collapse
Affiliation(s)
- Irene Ramos
- Department of Neurology and Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Konstantinos Stamatakis
- Centro de Biología Molecular Severo Ochoa, UAM-CSIC. Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; (K.S.); (C.L.O.)
| | - Clara L. Oeste
- Centro de Biología Molecular Severo Ochoa, UAM-CSIC. Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain; (K.S.); (C.L.O.)
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu, 9, 28040 Madrid, Spain
| |
Collapse
|
41
|
Jang B, Kim M, Lee Y, Ishigami A, Kim Y, Choi E. Vimentin citrullination probed by a novel monoclonal antibody serves as a specific indicator for reactive astrocytes in neurodegeneration. Neuropathol Appl Neurobiol 2020; 46:751-769. [DOI: 10.1111/nan.12620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/23/2020] [Indexed: 01/11/2023]
Affiliation(s)
- B. Jang
- Ilsong Institute of Life Science Hallym University Anyang Gyeonggi‐doRepublic of Korea
| | - M.J. Kim
- Ilsong Institute of Life Science Hallym University Anyang Gyeonggi‐doRepublic of Korea
- Department of Biomedical Gerontology Graduate School of Hallym University Chuncheon Gangwon‐do Republic of Korea
| | - Y.J. Lee
- Ilsong Institute of Life Science Hallym University Anyang Gyeonggi‐doRepublic of Korea
| | - A. Ishigami
- Molecular Regulation of Aging Tokyo Metropolitan Institute of Gerontology Itabashi‐ku Tokyo Japan
| | - Y.S. Kim
- Ilsong Institute of Life Science Hallym University Anyang Gyeonggi‐doRepublic of Korea
- Department of Microbiology College of Medicine Hallym University Chuncheon Gangwon‐do Republic of Korea
| | - E.K. Choi
- Ilsong Institute of Life Science Hallym University Anyang Gyeonggi‐doRepublic of Korea
- Department of Biomedical Gerontology Graduate School of Hallym University Chuncheon Gangwon‐do Republic of Korea
| |
Collapse
|
42
|
Bacterial type III effector protein HopQ inhibits melanoma motility through autophagic degradation of vimentin. Cell Death Dis 2020; 11:231. [PMID: 32286254 PMCID: PMC7156461 DOI: 10.1038/s41419-020-2427-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
Malignant melanoma is a fatal disease that rapidly spreads to the whole body. Treatments have limited efficiency owing to drug resistance and various side effects. Pseudomonas syringae pv. tomato (Pto) is a model bacterial pathogen capable of systemic infection in plants. Pto injects the effector protein HopQ into the plant cytosol via a type III secretion machinery and suppresses the host immunity. Intriguingly, host plant proteins regulated by HopQ are conserved even in humans and conferred in tumor metastasis. Nevertheless, the potential for HopQ to regulate human cancer metastasis was unknown. In this study, we addressed the suitability of HopQ as a possible drug against melanoma metastasis. In melanoma cells, overexpressed HopQ is phosphorylated and bound to 14-3-3 through its N-terminal domain, resulting in stronger interaction between HopQ and vimentin. The binding of HopQ to vimentin allowed for degradation of vimentin via p62-dependent selective autophagy. Attenuation of vimentin expression by HopQ inhibited melanoma motility and in vivo metastasis. These findings demonstrated that HopQ directly degraded vimentin in melanoma cells and could be applied to an inhibitor of melanoma metastasis.
Collapse
|
43
|
Wu X, Li J, Gassa A, Buchner D, Alakus H, Dong Q, Ren N, Liu M, Odenthal M, Stippel D, Bruns C, Zhao Y, Wahba R. Circulating tumor DNA as an emerging liquid biopsy biomarker for early diagnosis and therapeutic monitoring in hepatocellular carcinoma. Int J Biol Sci 2020; 16:1551-1562. [PMID: 32226301 PMCID: PMC7097921 DOI: 10.7150/ijbs.44024] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
As one of the most common malignant tumors worldwide, hepatocellular carcinoma (HCC) is known for its poor prognosis due to diagnosis only in advanced stages. Nearly 50% of the patients with the first diagnosis of HCC die within a year. Currently, the advancements in the integration of omics information have begun to transform the clinical management of cancer patients. Molecular profiling for HCC patients is in general obtained from resected tumor materials or biopsies. However, the resected tumor tissue is limited and can only be obtained through surgery, so that dynamic monitoring of patients cannot be performed. Compared to invasive procedures, circulating tumor DNA (ctDNA) has been proposed as an alternative source to perform molecular profiling of tumor DNA in cancer patients. The detection of abnormal forms of circulating cell-free DNA (cfDNA) that originate from cancer cells (ctDNA) provides a novel tool for cancer detection and disease monitoring. This may also be an opportunity to optimize the early diagnosis of HCC. In this review, we summarized the updated methods, materials, storage of sampling, detection techniques for ctDNA and the comparison of the applications among different biomarkers in HCC patients. In particular, we analyzed ctDNA studies dealing with copy number variations, gene integrity, mutations (RAS, TERT, CTNNB1, TP53 and so on), DNA methylation alterations (DBX2, THY1, TGR5 and so on) for the potential utility of ctDNA in the diagnosis and management of HCC. The biological functions and correlated signaling pathways of ctDNA associated genes (including MAPK/RAS pathway, p53 signaling pathway and Wnt-β catenin pathway) are also discussed and highlighted. Thus, exploration of ctDNA/cfDNA as potential biomarkers may provide a great opportunity in future liquid biopsy applications for HCC.
Collapse
Affiliation(s)
- Xiaolin Wu
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Jiahui Li
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Asmae Gassa
- Department of Cardiothoracic Surgery, Heart Center, University Hospital of Cologne, Germany, Kerpener Straße 62, 5.937 Cologne, Germany
| | - Denise Buchner
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, 200032, Shanghai, P.R. China
| | - Ning Ren
- Liver Cancer Institute & Zhongshan Hospital; Department of Surgery, Institute of Fudan-Minhang Academic Health System, Minhang Branch, Zhongshan Hospital, Fudan University, 200032, Shanghai, P.R. China
| | - Ming Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, 510095, Guangzhou, P.R. China
| | - Margarete Odenthal
- Institute of Pathology, University Hospital of Cologne, 50937, Cologne, Germany
| | - Dirk Stippel
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
- Department of General, Visceral und Vascular Surgery, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Roger Wahba
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| |
Collapse
|
44
|
Abstract
Caspases are a family of conserved cysteine proteases that play key roles in programmed cell death and inflammation. In multicellular organisms, caspases are activated via macromolecular signaling complexes that bring inactive procaspases together and promote their proximity-induced autoactivation and proteolytic processing. Activation of caspases ultimately results in programmed execution of cell death, and the nature of this cell death is determined by the specific caspases involved. Pioneering new research has unraveled distinct roles and cross talk of caspases in the regulation of programmed cell death, inflammation, and innate immune responses. In-depth understanding of these mechanisms is essential to foster the development of precise therapeutic targets to treat autoinflammatory disorders, infectious diseases, and cancer. This review focuses on mechanisms governing caspase activation and programmed cell death with special emphasis on the recent progress in caspase cross talk and caspase-driven gasdermin D-induced pyroptosis.
Collapse
Affiliation(s)
- Sannula Kesavardhana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; , ,
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; , ,
| | | |
Collapse
|
45
|
Siciliano RA, Mazzeo MF, Ferretta A, Pacelli C, Rosato A, Papa F, Scacco S, Papa S, Cocco T, Lippolis R. Decreased amount of vimentin N-terminal truncated proteolytic products in parkin-mutant skin fibroblasts. Biochem Biophys Res Commun 2020; 521:693-698. [PMID: 31699368 DOI: 10.1016/j.bbrc.2019.10.154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/22/2019] [Indexed: 10/25/2022]
Abstract
Vimentin, a member of cytoskeleton intermediate filaments proteins, plays a critical role in cell structure and dynamics. The present proteomic study reveals reduced amount of six different lengths, N-terminal truncated proteolytic products of vimentin, in the primary skin fibroblasts from two unrelated PD patients, as compared to control fibroblasts. The decreased amount of N-terminal truncated forms of vimentin in parkin-mutant fibroblasts, could contribute to impairment of cellular function, potentially contributing to the pathogenesis of Parkinson disease.
Collapse
Affiliation(s)
| | | | - Anna Ferretta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs University of Bari "Aldo Moro" Bari, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Rosato
- Department of Chemistry, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Papa
- Department of Basic Medical Sciences, Neurosciences and Sense Organs University of Bari "Aldo Moro" Bari, Italy
| | - Salvatore Scacco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs University of Bari "Aldo Moro" Bari, Italy
| | - Sergio Papa
- Department of Basic Medical Sciences, Neurosciences and Sense Organs University of Bari "Aldo Moro" Bari, Italy; Stazione Zoologica Anton Dohrn, Napoli, Italy
| | - Tiziana Cocco
- Department of Basic Medical Sciences, Neurosciences and Sense Organs University of Bari "Aldo Moro" Bari, Italy.
| | - Rosa Lippolis
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Bari, Italy.
| |
Collapse
|
46
|
Vimentin modulates apoptosis and inflammatory cytokine release by a human monocytic cell line (THP-1) in response to lipopolysaccharides in vitro. Chin Med J (Engl) 2019; 132:1336-1343. [PMID: 30882463 PMCID: PMC6629360 DOI: 10.1097/cm9.0000000000000187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND It has recently been recognized that serum vimentin is elevated in infectious diseases, and that vimentin plays a role in regulating neutrophils and macrophages associated inflammation. However, the mechanisms are unclear. This study was designed to explore the role of vimentin in regulating monocyte survival or apoptosis as well as inflammatory cytokine secretion in response to lipopolysaccharides (LPSs). METHODS A human monocytic leukemia cell line (THP-1) was transfected with vimentin-specific small interfering RNA (siRNA) or vimentin over-expressing plasmid. Apoptosis was assessed by TdT-mediated dUTP Nick-End Labeling (TUNEL) and DNA content assay. Immunoblotting was performed to detect apoptosis-associated proteins. Cytokines (interleukin [IL]-6, IL-10, and tumor necrosis factor α [TNF-α]) were measured by enzyme-linked immuno sorbent assay. Two-way analysis of variance followed by Student's t test was used to compare means between different groups. RESULTS Suppression of vimentin in THP-1 cells resulted in increased apoptotic response in the presence of LPS, while over-expression of vimentin could prevent the cells from apoptosis in response to LPS. LPS alone or suppression of vimentin resulted in significant up-regulation of caspase-3 (1.42 ± 0.20 of LPS alone and 1.68 ± 0.10 of vimentin suppression vs. control, t = 5.21 and 10.28, respectively, P < 0.05). In addition, pro-inflammatory cytokines (IL-6 and TNF-α) was significantly increased (IL-6: 577.90 ± 159.90 pg/day/10 cells vs. 283.80 ± 124.60 pg/day/10 cells of control, t = 14.76, P < 0.05; TNF-α: 54.10 ± 5.80 vs. 17.10 ± 0.10 pg/day/10 cells of control, t = 6.71, P < 0.05), while anti-inflammatory cytokine (IL-10) was significantly up-regulated in the THP-1 cells that over-expressed vimentin (140.9 ± 17.2 pg/day/10 cells vs. undetectable in control cells). CONCLUSIONS In summary, the vimentin may regulate innate immunity through modulating monocytes viability as well as inflammatory response in sepsis through shifting the balance of pro-inflammatory and anti-inflammatory cytokines.
Collapse
|
47
|
The Cytoskeleton as Regulator of Cell Signaling Pathways. Trends Biochem Sci 2019; 45:96-107. [PMID: 31812462 DOI: 10.1016/j.tibs.2019.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
During interphase, filamentous actin, microtubules, and intermediate filaments regulate cell shape, motility, transport, and interactions with the environment. These activities rely on signaling events that control cytoskeleton properties. Recent studies uncovered mechanisms that go far beyond this one-directional flow of information. Thus, the three branches of the cytoskeleton impinge on signaling pathways to determine their activities. We propose that this regulatory role of the cytoskeleton provides sophisticated mechanisms to control the spatiotemporal output and the intensity of signaling events. Specific examples emphasize these emerging contributions of the cytoskeleton to cell physiology. In our opinion, further exploration of these pathways will uncover new concepts of cellular communication that originate from the cytoskeleton.
Collapse
|
48
|
Gagliardi PA, Primo L. Death for life: a path from apoptotic signaling to tissue-scale effects of apoptotic epithelial extrusion. Cell Mol Life Sci 2019; 76:3571-3581. [PMID: 31143959 PMCID: PMC11105432 DOI: 10.1007/s00018-019-03153-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 12/24/2022]
Abstract
Apoptosis plays a crucial role in clearing old or critically compromised cells, and actively maintains epithelial homeostasis and epithelial morphogenesis during embryo development. But how is the apoptotic signaling pathway able to orchestrate such complex and dynamic multi-cellular morphological events at the tissue scale? In this review we collected the most updated knowledge regarding how apoptosis controls different cytoskeletal components. We describe how apoptosis can control epithelial homeostasis though epithelial extrusion, a highly orchestrated process based on high- order actomyosin structures and on the coordination between the apoptotic and the neighboring cells. Finally, we describe how the synergy among forces generated by multiple apoptotic cells can shape epithelia in embryo development.
Collapse
Affiliation(s)
- Paolo Armando Gagliardi
- Candiolo Cancer Institute-FPO IRCCS, 10060, Candiolo, Italy
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012, Bern, Switzerland
| | - Luca Primo
- Candiolo Cancer Institute-FPO IRCCS, 10060, Candiolo, Italy.
- Department of Oncology, University of Torino, 10060, Turin, Italy.
| |
Collapse
|
49
|
Pradeepkiran JA. Insights of rheumatoid arthritis risk factors and associations. J Transl Autoimmun 2019; 2:100012. [PMID: 32743500 PMCID: PMC7388374 DOI: 10.1016/j.jtauto.2019.100012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a defective post-translational modification of citrullinated peptides which cause synovial inflammation in joints. The present review elaborates the basic mechanisms of RA and the root causes of molecular mechanisms. The gender-based differentiation and probabilitiesof RA causes were discussed. Many report studies supporting that females are more prone to RA than males maybe suspected that circulating estrogen hormones 16a-hydroxy estrone, 2-hydroxy estrogens involvement in the RA pathogenicity. Other important aspects like environmental factors and air pollutants like (SO2 and NO2) were also impacted and enhances the risk of RA were discussed. The root cause of pathomechanisms of peptidylarginine deiminase (PAD) enzymes in RA and autoimmunity factors were poorly understood, however, Ati-citrullinated peptides (ACP) are the powerful markers to diagnose the RA disease. This review discusses three main risk factors of RA to understand the RA pathogenesis and disease-modifying mechanisms, may provide a unique opportunity to determine disease prevalence and RA associations. Rheumatoid arthritis(RA) is caused by the defected peptides, environmental factor’s and hormonal dysregulation in synovial inflammation. The immune system attacks joint tissue for pathogenic citrullinated peptides causing inflammation in synovium, leading to RA. Risk factors, disorder proteins, cellular changes influence immune system to turns to self antigens leads to RA. Understanding the exact role and action of risk factors in RA is especially important given the prevention measures to RA are desirable.
Collapse
Affiliation(s)
- Jangampalli Adi Pradeepkiran
- Sri Venkateswara University, Tirupati, 517502, AP, India
- Deprtment of Internal Medicine, Texas Tech University of Health Science Centre, Lubbock, USA
- Corresponding author. Department of Internal Medicine, Texas Tech University of Health Science Centre, Lubbock, USA.
| |
Collapse
|
50
|
Wenta T, Rychlowski M, Jurewicz E, Jarzab M, Zurawa-Janicka D, Filipek A, Lipinska B. The HtrA3 protease promotes drug-induced death of lung cancer cells by cleavage of the X-linked inhibitor of apoptosis protein (XIAP). FEBS J 2019; 286:4579-4596. [PMID: 31260151 DOI: 10.1111/febs.14977] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/09/2019] [Accepted: 06/27/2019] [Indexed: 01/02/2023]
Abstract
HtrA3 is a proapoptotic protease shown to promote drug-induced cytotoxicity in lung cancer cells and proposed to have an antitumor effect. However, at the molecular level, the role of HtrA3 in cell death induction is poorly understood. There are two HtrA3 isoforms, a long and a short one, termed HtrA3L and HtrA3S. By performing pull down assays, co-immunoprecipitation and ELISA, we showed that HtrA3 formed complexes with the X-linked inhibitor of apoptosis protein (XIAP). The recombinant HtrA3 variants ΔN-HtrA3L and -S, lacking the N-terminal regions that are not essential for protease activity, cleaved XIAP with a comparable efficiency, though ΔN-HtrA3S was more active in the presence of cellular extract, suggesting the existence of an activating factor. Immunofluorescence and proximity ligation assays indicated that HtrA3 partially co-localized with XIAP. Exogenous ΔN-HtrA3L/S promoted apoptotic death of lung cancer cells treated with etoposide and caused a significant decrease of cellular XIAP levels, in a way dependent on HtrA3 proteolytic activity. These results collectively indicate that both HtrA3 isoforms stimulate drug-induced apoptotic death of lung cancer cells via XIAP cleavage and thus help to understand the molecular mechanism of HtrA3 function in apoptosis and in cancer cell death caused by chemotherapy.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Michal Rychlowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk - Medical University of Gdansk, Poland
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| |
Collapse
|