51
|
Matusali G, Trionfetti F, Bordoni V, Nardacci R, Falasca L, Colombo D, Terri M, Montaldo C, Castilletti C, Mariotti D, Del Nonno F, Capobianchi MR, Agrati C, Tripodi M, Strippoli R. Pleural Mesothelial Cells Modulate the Inflammatory/Profibrotic Response During SARS-CoV-2 Infection. Front Mol Biosci 2021; 8:752616. [PMID: 34901152 PMCID: PMC8662383 DOI: 10.3389/fmolb.2021.752616] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022] Open
Abstract
Although lung fibrosis has a major impact in COVID-19 disease, its pathogenesis is incompletely understood. In particular, no direct evidence of pleura implication in COVID-19-related fibrotic damage has been reported so far. In this study, the expression of epithelial cytokeratins and Wilms tumor 1 (WT1), specific markers of mesothelial cells (MCs), was analyzed in COVID-19 and unrelated pleura autoptic samples. SARS-CoV-2 replication was analyzed by RT-PCR and confocal microscopy in MeT5A, a pleura MC line. SARS-CoV-2 receptors were analyzed by RT-PCR and western blot. Inflammatory cytokines from the supernatants of SARS-CoV-2-infected MeT5A cells were analysed by Luminex and ELLA assays. Immunohistochemistry of COVID-19 pleura patients highlighted disruption of pleura monolayer and fibrosis of the sub-mesothelial stroma, with the presence of MCs with fibroblastoid morphology in the sub-mesothelial stroma, but no evidence of direct infection in vivo. Interestingly, we found evidence of ACE2 expression in MCs from pleura of COVID-19 patients. In vitro analysis shown that MeT5A cells expressed ACE2, TMPRSS2, ADAM17 and NRP1, plasma membrane receptors implicated in SARS-CoV-2 cell entry and infectivity. Moreover, MeT5A cells sustained SARS-CoV-2 replication and productive infection. Infected MeT5A cells produced interferons, inflammatory cytokines and metalloproteases. Overall, our data highlight the potential role of pleura MCs as promoters of the fibrotic reaction and regulators of the immune response upon SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Veronica Bordoni
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Roberta Nardacci
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy.,UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Laura Falasca
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Daniele Colombo
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Concetta Castilletti
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Davide Mariotti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Franca Del Nonno
- Laboratory of Electron Microscopy, National Institute for Infectious Diseases "Lazzaro Spallanzani", IRCCS, Rome, Italy
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Chiara Agrati
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.,Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
52
|
Soliman F, Ye L, Jiang W, Hargest R. Targeting Hyaluronic Acid and Peritoneal Dissemination in Colorectal Cancer. Clin Colorectal Cancer 2021; 21:e126-e134. [PMID: 34955378 DOI: 10.1016/j.clcc.2021.11.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/30/2021] [Accepted: 11/22/2021] [Indexed: 11/03/2022]
Abstract
Peritoneal metastasis (PM) from colorectal cancer (CRC) carries a significant mortality rate for patients and treatment is challenging. The development of PM is a multistep process involving detachment, adhesion, invasion and colonization of the peritoneal cavity. Cytoreductive surgery and HIPEC (hyperthermic intraperitoneal chemotherapy) for PM from CRC has some benefit but overall survival is poor and recurrence rates are high. Treatments to prevent the development of peritoneal metastasis could have the potential to improve CRC survival and disease-free outcomes. The ability of cancer cells to invade the peritoneum and become established as metastatic tumors is influenced by a multifactorial process. Hyaluronic acid (HA) has been shown to coat the mesothelial cells of the peritoneum and has been demonstrated to be utilized in various malignancies as part of the metastatic process in peritoneal dissemination. CD44, RHAMM (CD168) and ICAM-1 have all been shown to be binding partners for HA. Targeting HA-mediated binding may prevent adhesion to distant sites within the peritoneum through suppression of interaction of these molecules. Here we review the current literature and discuss key molecules involved with PM dissemination, with the potential to target these mechanisms in the delivery of future treatments.
Collapse
Affiliation(s)
- Faris Soliman
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University; Cardiff and Vale University Health Board.
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University
| | - Rachel Hargest
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, School of Medicine, Cardiff University; Cardiff and Vale University Health Board
| |
Collapse
|
53
|
Cui H, Liu G. DOCK-t(w)o Pleural Fibrosis. Am J Respir Cell Mol Biol 2021; 66:117-119. [PMID: 34758277 PMCID: PMC8845136 DOI: 10.1165/rcmb.2021-0411ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Huachun Cui
- University of alabama at birmingham, Birmingham, Alabama, United States
| | - Gang Liu
- University of Alabama at Birmingham, Birmingham, Alabama, United States;
| |
Collapse
|
54
|
Resident macrophage-dependent immune cell scaffolds drive anti-bacterial defense in the peritoneal cavity. Immunity 2021; 54:2578-2594.e5. [PMID: 34717795 DOI: 10.1016/j.immuni.2021.10.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/13/2021] [Accepted: 10/11/2021] [Indexed: 12/25/2022]
Abstract
Peritoneal immune cells reside unanchored within the peritoneal fluid in homeostasis. Here, we examined the mechanisms that control bacterial infection in the peritoneum using a mouse model of abdominal sepsis following intraperitoneal Escherichia coli infection. Whole-mount immunofluorescence and confocal microscopy of the peritoneal wall and omentum revealed that large peritoneal macrophages (LPMs) rapidly cleared bacteria and adhered to the mesothelium, forming multilayered cellular aggregates composed by sequentially recruited LPMs, B1 cells, neutrophils, and monocyte-derived cells (moCs). The formation of resident macrophage aggregates (resMφ-aggregates) required LPMs and thrombin-dependent fibrin polymerization. E. coli infection triggered LPM pyroptosis and release of inflammatory mediators. Resolution of these potentially inflammatory aggregates required LPM-mediated recruitment of moCs, which were essential for fibrinolysis-mediated resMφ-aggregate disaggregation and the prevention of peritoneal overt inflammation. Thus, resMφ-aggregates provide a physical scaffold that enables the efficient control of peritoneal infection, with implications for antimicrobial immunity in other body cavities, such as the pleural cavity or brain ventricles.
Collapse
|
55
|
Sommerfeld L, Finkernagel F, Jansen JM, Wagner U, Nist A, Stiewe T, Müller‐Brüsselbach S, Sokol AM, Graumann J, Reinartz S, Müller R. The multicellular signalling network of ovarian cancer metastases. Clin Transl Med 2021; 11:e633. [PMID: 34841720 PMCID: PMC8574964 DOI: 10.1002/ctm2.633] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/08/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transcoelomic spread is the major route of metastasis of ovarian high-grade serous carcinoma (HGSC) with the omentum as the major metastatic site. Its unique tumour microenvironment with its large populations of adipocytes, mesothelial cells and immune cells establishes an intercellular signaling network that is instrumental for metastatic growth yet poorly understood. METHODS Based on transcriptomic analysis of tumour cells, tumour-associated immune and stroma cells we defined intercellular signaling pathways for 284 cytokines and growth factors and their cognate receptors after bioinformatic adjustment for contaminating cell types. The significance of individual components of this network was validated by analysing clinical correlations and potentially pro-metastatic functions, including tumour cell migration, pro-inflammatory signal transduction and TAM expansion. RESULTS The data show an unexpected prominent role of host cells, and in particular of omental adipocytes, mesothelial cells and fibroblasts (CAF), in sustaining this signaling network. These cells, rather than tumour cells, are the major source of most cytokines and growth factors in the omental microenvironment (n = 176 vs. n = 13). Many of these factors target tumour cells, are linked to metastasis and are associated with a short survival. Likewise, tumour stroma cells play a major role in extracellular-matrix-triggered signaling. We have verified the functional significance of our observations for three exemplary instances. We show that the omental microenvironment (i) stimulates tumour cell migration and adhesion via WNT4 which is highly expressed by CAF; (ii) induces pro-tumourigenic TAM proliferation in conjunction with high CSF1 expression by omental stroma cells and (iii) triggers pro-inflammatory signaling, at least in part via a HSP70-NF-κB pathway. CONCLUSIONS The intercellular signaling network of omental metastases is majorly dependent on factors secreted by immune and stroma cells to provide an environment that supports ovarian HGSC progression. Clinically relevant pathways within this network represent novel options for therapeutic intervention.
Collapse
Affiliation(s)
- Leah Sommerfeld
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Julia M. Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological EndocrinologyUniversity Hospital (UKGM)MarburgGermany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological EndocrinologyUniversity Hospital (UKGM)MarburgGermany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- Institute of Molecular OncologyPhilipps UniversityMarburgGermany
| | - Sabine Müller‐Brüsselbach
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Anna M. Sokol
- The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Johannes Graumann
- The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Institute for Translational Proteomics, Philipps UniversityMarburgGermany
| | - Silke Reinartz
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| |
Collapse
|
56
|
Chua JW, Madden L, Lim SBH, Philips ARJ, Becker DL. Development of a refined ex vivo model of peritoneal adhesion formation, and a role for connexin 43 in their development. Mol Cell Biochem 2021; 477:295-305. [PMID: 34716547 DOI: 10.1007/s11010-021-04282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022]
Abstract
Despite many advances across the surgical sciences, post-surgical peritoneal adhesions still pose a considerable risk in modern-day procedures and are highly undesirable. We have developed a novel mouse peritoneal strip ex vivo adhesion model which may serve to bridge the gap between single cell culture systems and in vivo animal drug testing for the assessment of potential anti-adhesion agents, and study of causality of the process. We investigated the optimal conditions for adhesion formation with mouse peritoneal tissue strips by modifying an existing ex vivo rat model of peritoneal adhesions. We assessed the impact of the following conditions on the formation of adhesions: contact pressure, abrasions, and the presence of clotted blood. Macroscopic adhesions were detected in all mouse peritoneal strips exposed to specific conditions, namely abrasions and clotted blood, where peritoneal surfaces were kept in contact with pressure using cotton gauze in a tissue cassette. Adhesions were confirmed microscopically. Interestingly, connexin 43, a gap junction protein, was found to be upregulated at sites of adhesions. Key features of this model were the use of padding the abraded tissue with gauze and the use of a standardised volume of clotted blood. Using this model, peritoneal strips cultured with clotted blood between abraded surfaces were found to reproducibly develop adhesion bands at 72 h. Our goal is to develop a model that can be used in genetically modified mice in order to dissect out the role of particular genes in adhesion formation and to test drugs to prevent adhesion formation.
Collapse
Affiliation(s)
- Jia Wang Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore, 308232, Singapore
| | - Leigh Madden
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore, 308232, Singapore
| | - Sophia Beng Hui Lim
- Skin Research Institute Singapore, Level 17, Clinical Sciences Building, 11, Mandalay Road, Singapore, 308232, Singapore
| | | | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, Singapore, 308232, Singapore. .,Skin Research Institute Singapore, Level 17, Clinical Sciences Building, 11, Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
57
|
Qian G, Adeyanju O, Roy S, Sunil C, Jeffers A, Guo X, Ikebe M, Idell S, Tucker TA. DOCK2 Promotes Pleural Fibrosis by Modulating Mesothelial to Mesenchymal Transition. Am J Respir Cell Mol Biol 2021; 66:171-182. [PMID: 34710342 DOI: 10.1165/rcmb.2021-0175oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mesothelial to mesenchymal transition (MesoMT) is one of the crucial mechanisms underlying pleural fibrosis, which results in restrictive lung disease. DOCK2 plays important roles in immune functions, however, its role in pleural fibrosis particularly MesoMT remains unknown. We found that DOCK2 and the MesoMT maker α-SMA were significantly elevated and colocalized in the thickened pleura of patients with nonspecific pleuritis, suggesting the involvement of DOCK2 in the pathogenesis of MesoMT and pleural fibrosis. Likewise, data from three different pleural fibrosis models (TGF-β, carbon black/bleomycin, and streptococcal empyema) consistently demonstrated DOCK2 upregulation and its colocalization with α-SMA in the pleura. In addition, induced DOCK2 colocalized with the mesothelial marker calretinin, implicating DOCK2 in the regulation of MesoMT. Our in vivo data also showed that DOCK2 knockout mice were protected from Streptococcus pneumoniae induced pleural fibrosis, impaired lung compliance, and collagen deposition. To determine the involvement of DOCK2 in MesoMT, we treated primary human pleural mesothelial cells with the potent MesoMT inducer TGF-β. TGF-β significantly induced DOCK2 expression in a time-dependent manner, along with α-SMA, collagen 1, and fibronectin. Furthermore, DOCK2 knockdown significantly attenuated TGF-β induced α-SMA, collagen 1 and fibronectin expression, suggesting the importance of DOCK2 in TGF-β induced MesoMT. DOCK2 knockdown also inhibited TGF-β induced Snail upregulation, which may account for its role in regulating MesoMT. Taken together, the current study provides evidence that DOCK2 contributes to the pathogenesis of pleural fibrosis by mediating MesoMT and deposition of neomatrix and may represent a novel target for its prevention or treatment.
Collapse
Affiliation(s)
- Guoqing Qian
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States;
| | - Oluwaseun Adeyanju
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Saptarshi Roy
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Christudas Sunil
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Ann Jeffers
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Xia Guo
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Mitsuo Ikebe
- The University of Texas Health Science Center at Tyler, 12341, Department of Cellular and Molecular Biology, Tyler, Texas, United States
| | - Steven Idell
- The University of Texas Health Science Center at Tyler, 12341, Texas Lung Injury Institute, Tyler, Texas, United States
| | - Torry A Tucker
- The University of Texas Health Science Center at Tyler, 12341, Texas Lung Injury Institute, Tyler, Texas, United States
| |
Collapse
|
58
|
Progatzky F, Shapiro M, Chng SH, Garcia-Cassani B, Classon CH, Sevgi S, Laddach A, Bon-Frauches AC, Lasrado R, Rahim M, Amaniti EM, Boeing S, Shah K, Entwistle LJ, Suárez-Bonnet A, Wilson MS, Stockinger B, Pachnis V. Regulation of intestinal immunity and tissue repair by enteric glia. Nature 2021; 599:125-130. [PMID: 34671159 DOI: 10.1038/s41586-021-04006-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 09/07/2021] [Indexed: 02/06/2023]
Abstract
Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.
Collapse
Affiliation(s)
- Fränze Progatzky
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.
| | - Michael Shapiro
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.,AhRimmunity Laboratory, The Francis Crick Institute, London, UK
| | - Song Hui Chng
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.,Roche Innovation Center Shanghai, Shanghai, China
| | - Bethania Garcia-Cassani
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Cajsa Helena Classon
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Selin Sevgi
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Anna Laddach
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Ana Carina Bon-Frauches
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.,Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Reena Lasrado
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Maryam Rahim
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK
| | - Eleni-Maria Amaniti
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.,AhRimmunity Laboratory, The Francis Crick Institute, London, UK.,Sainsbury Wellcome Centre, London, UK
| | - Stefan Boeing
- Bioinformatics & Biostatistics STP, The Francis Crick Institute, London, UK
| | - Kathleen Shah
- AhRimmunity Laboratory, The Francis Crick Institute, London, UK
| | - Lewis J Entwistle
- AhRimmunity Laboratory, The Francis Crick Institute, London, UK.,Adaptive Immunity Research Unit, GSK, Stevenage, UK
| | - Alejandro Suárez-Bonnet
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, Hatfield, UK.,Experimental Histopathology STP, The Francis Crick Institute, London, UK
| | - Mark S Wilson
- Immunology Discovery, Genentech Inc, South San Francisco, CA, USA
| | | | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
59
|
Ramírez-Pavez TN, Martínez-Esparza M, Ruiz-Alcaraz AJ, Marín-Sánchez P, Machado-Linde F, García-Peñarrubia P. The Role of Peritoneal Macrophages in Endometriosis. Int J Mol Sci 2021; 22:ijms221910792. [PMID: 34639133 PMCID: PMC8509388 DOI: 10.3390/ijms221910792] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023] Open
Abstract
Endometriosis is an estrogen-dependent gynecological disorder, defined as the growth of endometrial stromal cells and glands at extrauterine sites. Endometriotic lesions are more frequently located into the abdominal cavity, although they can also be implanted in distant places. Among its etiological factors, the presence of immune dysregulation occupies a prominent place, pointing out the beneficial and harmful outcomes of macrophages in the pathogenesis of this disease. Macrophages are tissue-resident cells that connect innate and adaptive immunity, playing a key role in maintaining local homeostasis in healthy conditions and being critical in the development and sustainment of many inflammatory diseases. Macrophages accumulate in the peritoneal cavity of women with endometriosis, but their ability to clear migrated endometrial fragments seems to be inefficient. Hence, the characteristics of the peritoneal immune system in endometriosis must be further studied to facilitate the search for new diagnostic and therapeutic tools. In this review, we summarize recent relevant advances obtained in both mouse, as the main animal model used to study endometriosis, and human, focusing on peritoneal macrophages obtained from endometriotic patients and healthy donors, under the perspective of its future clinical translation to the role that these cells play on this pathology.
Collapse
Affiliation(s)
- Tamara N. Ramírez-Pavez
- Departamento de Bioquímica, Biología Molecular (B) e Inmunología, Facultad de Medicina, IMIB and Regional Campus of International Excellence “Campus Mare Nostrum”, Universidad de Murcia, 30100 Murcia, Spain; (T.N.R.-P.); (M.M.-E.); (A.J.R.-A.)
| | - María Martínez-Esparza
- Departamento de Bioquímica, Biología Molecular (B) e Inmunología, Facultad de Medicina, IMIB and Regional Campus of International Excellence “Campus Mare Nostrum”, Universidad de Murcia, 30100 Murcia, Spain; (T.N.R.-P.); (M.M.-E.); (A.J.R.-A.)
| | - Antonio J. Ruiz-Alcaraz
- Departamento de Bioquímica, Biología Molecular (B) e Inmunología, Facultad de Medicina, IMIB and Regional Campus of International Excellence “Campus Mare Nostrum”, Universidad de Murcia, 30100 Murcia, Spain; (T.N.R.-P.); (M.M.-E.); (A.J.R.-A.)
| | - Pilar Marín-Sánchez
- Servicio de Ginecología y Obstetricia, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB, 30120 Murcia, Spain;
| | - Francisco Machado-Linde
- Servicio de Ginecología y Obstetricia, Hospital Clínico Universitario Reina Sofía, CARM, 30002 Murcia, Spain;
| | - Pilar García-Peñarrubia
- Departamento de Bioquímica, Biología Molecular (B) e Inmunología, Facultad de Medicina, IMIB and Regional Campus of International Excellence “Campus Mare Nostrum”, Universidad de Murcia, 30100 Murcia, Spain; (T.N.R.-P.); (M.M.-E.); (A.J.R.-A.)
- Correspondence: ; Tel.: +34-8-6888-4673
| |
Collapse
|
60
|
Lovisa S. Epithelial-to-Mesenchymal Transition in Fibrosis: Concepts and Targeting Strategies. Front Pharmacol 2021; 12:737570. [PMID: 34557100 PMCID: PMC8454779 DOI: 10.3389/fphar.2021.737570] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT), an embryonic program relaunched during wound healing and in pathological conditions such as fibrosis and cancer, continues to gain the attention of the research community, as testified by the exponential trend of publications since its discovery in the seventies. From the first description as a mesenchymal transformation, the concept of EMT has been substantially refined as an in-depth comprehension of its functional role has recently emerged thanks to the implementation of novel mouse models as well as the use of sophisticated mathematical modeling and bioinformatic analysis. Nevertheless, attempts to targeting EMT in fibrotic diseases are at their infancy and continue to pose several challenges. The aim of this mini review is to recapitulate the most recent concepts in the EMT field and to summarize the different strategies which have been exploited to target EMT in fibrotic disorders.
Collapse
Affiliation(s)
- Sara Lovisa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.,IRCCS Humanitas Research Hospital, Rozzano (MI), Italy
| |
Collapse
|
61
|
Zheng Y, Pierce AF, Wagner WL, Khalil HA, Chen Z, Servais AB, Ackermann M, Mentzer SJ. Functional Adhesion of Pectin Biopolymers to the Lung Visceral Pleura. Polymers (Basel) 2021; 13:2976. [PMID: 34503016 PMCID: PMC8433721 DOI: 10.3390/polym13172976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/10/2023] Open
Abstract
Pleural injuries and the associated "air leak" are the most common complications after pulmonary surgery. Air leaks are the primary reason for prolonged chest tube use and increased hospital length of stay. Pectin, a plant-derived heteropolysaccharide, has been shown to be an air-tight sealant of pulmonary air leaks. Here, we investigate the morphologic and mechanical properties of pectin adhesion to the visceral pleural surface of the lung. After the application of high-methoxyl citrus pectin films to the murine lung, we used scanning electron microscopy to demonstrate intimate binding to the lung surface. To quantitatively assess pectin adhesion to the pleural surface, we used a custom adhesion test with force, distance, and time recordings. These assays demonstrated that pectin-glycocalyceal tensile adhesive strength was greater than nanocellulose fiber films or pressure-sensitive adhesives (p < 0.001). Simultaneous videomicroscopy recordings demonstrated that pectin-glycocalyceal adhesion was also stronger than the submesothelial connective tissue as avulsed surface remnants were visualized on the separated pectin films. Finally, pleural abrasion and hyaluronidase enzyme digestion confirmed that pectin binding was dependent on the pleural glycocalyx (p < 0.001). The results indicate that high methoxyl citrus pectin is a promising sealant for the treatment of pleural lung injuries.
Collapse
Affiliation(s)
- Yifan Zheng
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| | - Aidan F. Pierce
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| | - Willi L. Wagner
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
- Department of Diagnostic and Interventional Radiology, Translational Lung Research Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Hassan A. Khalil
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| | - Zi Chen
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| | - Andrew B. Servais
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany;
| | - Steven J. Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.Z.); (A.F.P.); (W.L.W.); (H.A.K.); (Z.C.); (A.B.S.)
| |
Collapse
|
62
|
Lee SM, Min YS, Son YK, Kim SE, An WS. Comparison of clinical outcome between incremental peritoneal dialysis and conventional peritoneal dialysis: a propensity score matching study. Ren Fail 2021; 43:1222-1228. [PMID: 34396922 PMCID: PMC8381909 DOI: 10.1080/0886022x.2021.1960564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background Incremental peritoneal dialysis (iPD) can be useful in patients with residual renal function (RRF). RRF was well preserved and similar survival was shown in iPD compared to conventional PD (cPD) in previous study. However, the long-term survival of iPD remains unclear compared to cPD in diabetic patients. This study evaluated whether patient survival, hospitalization and peritonitis, and PD survival in iPD were lower than cPD or not. Methods We conducted a 12-year retrospective observational study of 303 PD patients (232 cPD and 71 iPD) using propensity score matching by age, gender, and diabetes mellitus (DM). Finally, 78 cPD patients and 39 iPD patients were included and 44 patients had DM. Incremental PD was defined as starting PD with two or three manual exchanges per day. Results The median duration of iPD was 24.1 months and iPD had higher RRF than cPD. Compared to cPD, the patient survival, PD survival and hospitalization benefits were not found in iPD but diabetic iPD patients had significantly longer survival and less hospitalization. Cumulative risk for peritonitis was lower iPD and PD duration of iPD was longer than those of cPD. The iPD was an independent factor associated with survival in patients with DM. Conclusions Incremental PD may be a safe PD modality to initiate and maintain PD in less uremic patients with tolerable RRF. Incremental PD would be a benefit for survival in diabetic patients. Further prospective studies are necessary to confirm the effectiveness of iPD in PD patients with similar RRF.
Collapse
Affiliation(s)
- Su Mi Lee
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Yoon Sung Min
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Young Ki Son
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Seong Eun Kim
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
63
|
Wilm TP, Tanton H, Mutter F, Foisor V, Middlehurst B, Ward K, Benameur T, Hastie N, Wilm B. Restricted differentiative capacity of Wt1-expressing peritoneal mesothelium in postnatal and adult mice. Sci Rep 2021; 11:15940. [PMID: 34354169 PMCID: PMC8342433 DOI: 10.1038/s41598-021-95380-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 07/23/2021] [Indexed: 01/13/2023] Open
Abstract
Previously, genetic lineage tracing based on the mesothelial marker Wt1, appeared to show that peritoneal mesothelial cells have a range of differentiative capacities and are the direct progenitors of vascular smooth muscle in the intestine. However, it was not clear whether this was a temporally limited process or continued throughout postnatal life. Here, using a conditional Wt1-based genetic lineage tracing approach, we demonstrate that the postnatal and adult peritoneum covering intestine, mesentery and body wall only maintained itself and failed to contribute to other visceral tissues. Pulse-chase experiments of up to 6 months revealed that Wt1-expressing cells remained confined to the peritoneum and failed to differentiate into cellular components of blood vessels or other tissues underlying the peritoneum. Our data confirmed that the Wt1-lineage system also labelled submesothelial cells. Ablation of Wt1 in adult mice did not result in changes to the intestinal wall architecture. In the heart, we observed that Wt1-expressing cells maintained the epicardium and contributed to coronary vessels in newborn and adult mice. Our results demonstrate that Wt1-expressing cells in the peritoneum have limited differentiation capacities, and that contribution of Wt1-expressing cells to cardiac vasculature is based on organ-specific mechanisms.
Collapse
Affiliation(s)
- Thomas P Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Helen Tanton
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Fiona Mutter
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,ZIK Plasmatis "Plasma Redox Effects", Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Veronica Foisor
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Department of Chemistry, University of Warwick, Coventry, UK
| | - Ben Middlehurst
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Kelly Ward
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Tarek Benameur
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Department of Biomedical Sciences, College of Medicine, King Faisal University, Al Ahsa, Kingdom of Saudi Arabia
| | - Nicholas Hastie
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
64
|
Fatehi Hassanabad A, Zarzycki AN, Jeon K, Deniset JF, Fedak PWM. Post-Operative Adhesions: A Comprehensive Review of Mechanisms. Biomedicines 2021; 9:biomedicines9080867. [PMID: 34440071 PMCID: PMC8389678 DOI: 10.3390/biomedicines9080867] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/27/2021] [Accepted: 07/15/2021] [Indexed: 11/16/2022] Open
Abstract
Post-surgical adhesions are common in almost all surgical areas and are associated with significant rates of morbidity, mortality, and increased healthcare costs, especially when a patient requires repeat operative interventions. Many groups have studied the mechanisms driving post-surgical adhesion formation. Despite continued advancements, we are yet to identify a prevailing mechanism. It is highly likely that post-operative adhesions have a multifactorial etiology. This complex pathophysiology, coupled with our incomplete understanding of the underlying pathways, has resulted in therapeutic options that have failed to demonstrate safety and efficacy on a consistent basis. The translation of findings from basic and preclinical research into robust clinical trials has also remained elusive. Herein, we present and contextualize the latest findings surrounding mechanisms that have been implicated in post-surgical adhesion formation.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.F.D.)
| | - Anna N. Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.F.D.)
| | - Kristina Jeon
- Department of Anesthesiology and Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada;
| | - Justin F. Deniset
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.F.D.)
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Paul W. M. Fedak
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.F.D.)
- Correspondence:
| |
Collapse
|
65
|
Fatehi Hassanabad A, Zarzycki AN, Jeon K, Dundas JA, Vasanthan V, Deniset JF, Fedak PWM. Prevention of Post-Operative Adhesions: A Comprehensive Review of Present and Emerging Strategies. Biomolecules 2021; 11:biom11071027. [PMID: 34356652 PMCID: PMC8301806 DOI: 10.3390/biom11071027] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Post-operative adhesions affect patients undergoing all types of surgeries. They are associated with serious complications, including higher risk of morbidity and mortality. Given increased hospitalization, longer operative times, and longer length of hospital stay, post-surgical adhesions also pose a great financial burden. Although our knowledge of some of the underlying mechanisms driving adhesion formation has significantly improved over the past two decades, literature has yet to fully explain the pathogenesis and etiology of post-surgical adhesions. As a result, finding an ideal preventative strategy and leveraging appropriate tissue engineering strategies has proven to be difficult. Different products have been developed and enjoyed various levels of success along the translational tissue engineering research spectrum, but their clinical translation has been limited. Herein, we comprehensively review the agents and products that have been developed to mitigate post-operative adhesion formation. We also assess emerging strategies that aid in facilitating precision and personalized medicine to improve outcomes for patients and our healthcare system.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
| | - Anna N. Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
| | - Kristina Jeon
- Department of Anesthesiology and Pain Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada;
| | - Jameson A. Dundas
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
| | - Vishnu Vasanthan
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
| | - Justin F. Deniset
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Paul W. M. Fedak
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2N9, Canada; (A.F.H.); (A.N.Z.); (J.A.D.); (V.V.); (J.F.D.)
- Correspondence:
| |
Collapse
|
66
|
Masola V, Bonomini M, Onisto M, Ferraro PM, Arduini A, Gambaro G. Biological Effects of XyloCore, a Glucose Sparing PD Solution, on Mesothelial Cells: Focus on Mesothelial-Mesenchymal Transition, Inflammation and Angiogenesis. Nutrients 2021; 13:2282. [PMID: 34209455 PMCID: PMC8308380 DOI: 10.3390/nu13072282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Glucose-based solutions remain the most used osmotic agents in peritoneal dialysis (PD), but unavoidably they contribute to the loss of peritoneal filtration capacity. Here, we evaluated at a molecular level the effects of XyloCore, a new PD solution with a low glucose content, in mesothelial and endothelial cells. Cell viability, integrity of mesothelial and endothelial cell membrane, activation of mesothelial and endothelial to mesenchymal transition programs, inflammation, and angiogenesis were evaluated by several techniques. Results showed that XyloCore preserves mesothelial and endothelial cell viability and membrane integrity. Moreover XyloCore, unlike glucose-based solutions, does not exert pro-fibrotic, -inflammatory, and -angiogenic effects. Overall, the in vitro evidence suggests that XyloCore could represent a potential biocompatible solution promising better outcomes in clinical practice.
Collapse
Affiliation(s)
- Valentina Masola
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy;
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy;
| | - Pietro Manuel Ferraro
- U.O.S. Terapia Conservativa della Malattia Renale Cronica, U.O.C. Nefrologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00178 Rome, Italy;
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00178 Rome, Italy
| | - Arduino Arduini
- R&D Department, Iperboreal Pharma Srl, 65122 Pescara, Italy;
| | - Giovanni Gambaro
- Division of Nephrology and Dialysis, Department of Medicine, Piazzale A. Stefani 1, 37126 Verona, Italy;
| |
Collapse
|
67
|
Franklin RA. Fibroblasts and macrophages: Collaborators in tissue homeostasis. Immunol Rev 2021; 302:86-103. [PMID: 34101202 DOI: 10.1111/imr.12989] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022]
Abstract
Fibroblasts and macrophages are universal cell types across all mammalian tissues. These cells differ in many ways including their cellular origins; dynamics of renewal, recruitment, and motility within tissues; roles in tissue structure and secretion of signaling molecules; and contributions to the activation and progression of immune responses. However, many of the features that make these two cell types unique are not opposing, but instead complementary. This review will present cell-cell communication in this context and discuss how complementarity makes fibroblasts and macrophages highly compatible partners in the maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Ruth A Franklin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
68
|
Folliculin haploinsufficiency causes cellular dysfunction of pleural mesothelial cells. Sci Rep 2021; 11:10814. [PMID: 34031471 PMCID: PMC8144428 DOI: 10.1038/s41598-021-90184-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/07/2021] [Indexed: 11/10/2022] Open
Abstract
Birt–Hogg–Dubé syndrome (BHDS), an autosomal dominant inheritance disease caused by folliculin (FLCN) mutations, is associated with lung cysts and spontaneous pneumothorax. The possibility of FLCN haploinsufficiency in pleural mesothelial cells (PMCs) contributing to development of pneumothorax has not yet been clarified. Electron microscopy revealed exposed intercellular boundaries between PMCs on visceral pleura and decreased electron density around the adherens junctions in BHDS. To characterize cellular function of PMCs in BHDS patients (BHDS-PMCs), during surgery for pneumothorax, we established the flow cytometry-based methods of isolating high-purity PMCs from pleural lavage fluid. BHDS-PMCs showed impaired cell attachment and a significant decrease in proliferation and migration, but a significant increase in apoptosis compared with PMCs from primary spontaneous pneumothorax (PSP) patients (PSP-PMCs). Microarray analysis using isolated PMCs revealed a significant alteration in the expression of genes belonging to Gene Ontology terms “cell–cell adhesion junction” and “cell adhesion molecule binding”. Gene set enrichment analysis demonstrated that CDH1, encoding E-cadherin, was identified in the down-regulated leading edge of a plot in BHDS-PMCs. AMPK and LKB1 activation were significantly impaired in BHDS-PMCs compared with PSP-PMCs. Our findings indicate that FLCN haploinsufficiency may affect the E-cadherin-LKB1-AMPK axis and lead to abnormal cellular function in BHDS-PMCs.
Collapse
|
69
|
Zwicky SN, Stroka D, Zindel J. Sterile Injury Repair and Adhesion Formation at Serosal Surfaces. Front Immunol 2021; 12:684967. [PMID: 34054877 PMCID: PMC8160448 DOI: 10.3389/fimmu.2021.684967] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022] Open
Abstract
Most multicellular organisms have a major body cavity containing vital organs. This cavity is lined by a mucosa-like serosal surface and filled with serous fluid which suspends many immune cells. Injuries affecting the major body cavity are potentially life-threatening. Here we summarize evidence that unique damage detection and repair mechanisms have evolved to ensure immediate and swift repair of injuries at serosal surfaces. Furthermore, thousands of patients undergo surgery within the abdominal and thoracic cavities each day. While these surgeries are potentially lifesaving, some patients will suffer complications due to inappropriate scar formation when wound healing at serosal surfaces defects. These scars called adhesions cause profound challenges for health care systems and patients. Therefore, reviewing the mechanisms of wound repair at serosal surfaces is of clinical importance. Serosal surfaces will be introduced with a short embryological and microanatomical perspective followed by a discussion of the mechanisms of damage recognition and initiation of sterile inflammation at serosal surfaces. Distinct immune cells populations are free floating within the coelomic (peritoneal) cavity and contribute towards damage recognition and initiation of wound repair. We will highlight the emerging role of resident cavity GATA6+ macrophages in repairing serosal injuries and compare serosal (mesothelial) injuries with injuries to the blood vessel walls. This allows to draw some parallels such as the critical role of the mesothelium in regulating fibrin deposition and how peritoneal macrophages can aggregate in a platelet-like fashion in response to sterile injury. Then, we discuss how serosal wound healing can go wrong, causing adhesions. The current pathogenetic understanding of and potential future therapeutic avenues against adhesions are discussed.
Collapse
Affiliation(s)
- Simone N Zwicky
- Department of Visceral Surgery and Medicine, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Joel Zindel
- Department of Visceral Surgery and Medicine, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
70
|
Terri M, Trionfetti F, Montaldo C, Cordani M, Tripodi M, Lopez-Cabrera M, Strippoli R. Mechanisms of Peritoneal Fibrosis: Focus on Immune Cells-Peritoneal Stroma Interactions. Front Immunol 2021; 12:607204. [PMID: 33854496 PMCID: PMC8039516 DOI: 10.3389/fimmu.2021.607204] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Peritoneal fibrosis is characterized by abnormal production of extracellular matrix proteins leading to progressive thickening of the submesothelial compact zone of the peritoneal membrane. This process may be caused by a number of insults including pathological conditions linked to clinical practice, such as peritoneal dialysis, abdominal surgery, hemoperitoneum, and infectious peritonitis. All these events may cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy. Among the cellular processes implicated in these peritoneal alterations is the generation of myofibroblasts from mesothelial cells and other cellular sources that are central in the induction of fibrosis and in the subsequent functional deterioration of the peritoneal membrane. Myofibroblast generation and activity is actually integrated in a complex network of extracellular signals generated by the various cellular types, including leukocytes, stably residing or recirculating along the peritoneal membrane. Here, the main extracellular factors and the cellular players are described with emphasis on the cross-talk between immune system and cells of the peritoneal stroma. The understanding of cellular and molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.
Collapse
Affiliation(s)
- Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Claudia Montaldo
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marco Cordani
- instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA) Nanociencia, Madrid, Spain
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Manuel Lopez-Cabrera
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular “Severo Ochoa”-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
71
|
Kuper CF, Pieters RHH, van Bilsen JHM. Nanomaterials and the Serosal Immune System in the Thoracic and Peritoneal Cavities. Int J Mol Sci 2021; 22:ijms22052610. [PMID: 33807632 PMCID: PMC7961545 DOI: 10.3390/ijms22052610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/23/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
The thoracic and peritoneal cavities are lined by serous membranes and are home of the serosal immune system. This immune system fuses innate and adaptive immunity, to maintain local homeostasis and repair local tissue damage, and to cooperate closely with the mucosal immune system. Innate lymphoid cells (ILCs) are found abundantly in the thoracic and peritoneal cavities, and they are crucial in first defense against pathogenic viruses and bacteria. Nanomaterials (NMs) can enter the cavities intentionally for medical purposes, or unintentionally following environmental exposure; subsequent serosal inflammation and cancer (mesothelioma) has gained significant interest. However, reports on adverse effects of NM on ILCs and other components of the serosal immune system are scarce or even lacking. As ILCs are crucial in the first defense against pathogenic viruses and bacteria, it is possible that serosal exposure to NM may lead to a reduced resistance against pathogens. Additionally, affected serosal lymphoid tissues and cells may disturb adipose tissue homeostasis. This review aims to provide insight into key effects of NM on the serosal immune system.
Collapse
Affiliation(s)
- C. Frieke Kuper
- Consultant, Haagstraat 13, 3581 SW Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| | - Raymond H. H. Pieters
- Immunotoxicology, Institute for Risk Assessment Sciences, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands;
- Innovative Testing in Life Sciences & Chemistry, Research Centre for Healthy and Sustainable Living, University of Applied Sciences Utrecht, Padualaan 97, 3584 CH Utrecht, The Netherlands
| | - Jolanda H. M. van Bilsen
- Department for Risk Analysis for Products in Development, Netherlands Organization for Applied Scientific Research (TNO), Princetonlaan 6, 3584 CB Utrecht, The Netherlands
- Correspondence: (C.F.K.); (J.H.M.v.B.)
| |
Collapse
|
72
|
Chong WH, Saha BK, Austin A, Chopra A. The Significance of Subpleural Sparing in CT Chest: A State-of-the-Art Review. Am J Med Sci 2021; 361:427-435. [PMID: 33487401 DOI: 10.1016/j.amjms.2021.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/03/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022]
Abstract
The subpleural sparing pattern is a common finding on computed tomography (CT) of the lungs. It comprises of pulmonary opacities sparing the lung peripheries, typically 1cm and less from the pleural surface. This finding has a variety of causes, including idiopathic, inflammatory, infectious, inhalational, cardiac, traumatic, and bleeding disorders. Specific disorders that can cause subpleural sparing patterns include nonspecific interstitial pneumonia (NSIP), organizing pneumonia (OP), pulmonary alveolar proteinosis (PAP), diffuse alveolar hemorrhage (DAH), vaping-associated lung injury (VALI), cracked lung, pulmonary edema, pneumocystis jirovecii pneumonia (PJP), pulmonary contusion, and more recently, Coronavirus disease 2019 (COVID-19) pneumonia. Knowledge of the many etiologies of this pattern can be useful in preventing diagnostic errors. In addition, although the etiology of subpleural sparing pattern is frequently indistinguishable during an initial radiologic evaluation, the differences in location of opacities in the lungs, as well as the presence of additional radiologic findings, patient history, and clinical presentation, can often be useful to suggest the appropriate diagnosis. We did a comprehensive search on Pubmed and Google Scholar database using keywords of "subpleural sparing," "peripheral sparing," "sparing of peripheries," "CT chest," "chest imaging," and "pulmonary disease." This review aims to describe the primary differential diagnosis of subpleural sparing pattern seen on chest imaging with a strong emphasis on clinical and radiographic findings. We also discuss the pathogenesis and essential clues that are crucial to narrow the differential diagnosis.
Collapse
Affiliation(s)
- Woon H Chong
- Department of Pulmonary and Critical Care, Albany Medical Center, Albany, New York.
| | - Biplab K Saha
- Department of Pulmonary and Critical Care, Ozarks Medical Center, West Plains, Missouri
| | - Adam Austin
- Department of Pulmonary and Critical Care, University of Florida, Gainesville, Florida
| | - Amit Chopra
- Department of Pulmonary and Critical Care, Albany Medical Center, Albany, New York
| |
Collapse
|
73
|
Preparation of a cross-linked cartilage acellular matrix-poly (caprolactone-ran-lactide-ran-glycolide) film and testing its feasibility as an anti-adhesive film. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111283. [DOI: 10.1016/j.msec.2020.111283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/28/2020] [Accepted: 07/19/2020] [Indexed: 12/29/2022]
|
74
|
Calabrò ML, Lazzari N, Rigotto G, Tonello M, Sommariva A. Role of Epithelial-Mesenchymal Plasticity in Pseudomyxoma Peritonei: Implications for Locoregional Treatments. Int J Mol Sci 2020; 21:ijms21239120. [PMID: 33266161 PMCID: PMC7731245 DOI: 10.3390/ijms21239120] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanisms by which neoplastic cells disseminate from the primary tumor to metastatic sites, so-called metastatic organotropism, remain poorly understood. Epithelial-mesenchymal transition (EMT) plays a role in cancer development and progression by converting static epithelial cells into the migratory and microenvironment-interacting mesenchymal cells, and by the modulation of chemoresistance and stemness of tumor cells. Several findings highlight that pathways involved in EMT and its reverse process (mesenchymal-epithelial transition, MET), now collectively called epithelial-mesenchymal plasticity (EMP), play a role in peritoneal metastases. So far, the relevance of factors linked to EMP in a unique peritoneal malignancy such as pseudomyxoma peritonei (PMP) has not been fully elucidated. In this review, we focus on the role of epithelial-mesenchymal dynamics in the metastatic process involving mucinous neoplastic dissemination in the peritoneum. In particular, we discuss the role of expression profiles and phenotypic transitions found in PMP in light of the recent concept of EMP. A better understanding of EMP-associated mechanisms driving peritoneal metastasis will help to provide a more targeted approach for PMP patients selected for locoregional interventions involving cytoreductive surgery and hyperthermic intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
- Correspondence:
| | - Nayana Lazzari
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
| | - Giulia Rigotto
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (N.L.); (G.R.)
| | - Marco Tonello
- Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy;
| | - Antonio Sommariva
- Advanced Surgical Oncology, Surgical Oncology of the Esophagus and Digestive Tract, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy;
| |
Collapse
|
75
|
Papparella A, Noviello C, Ranucci S, Paciello O, Papparella S, De Biase D, Cirillo G, Umano GR. Pneumoperitoneum Modifies Serum and Tissue CCL2-CCL5 Expression in Mice. JSLS 2020; 24:JSLS.2020.00017. [PMID: 32508487 PMCID: PMC7242020 DOI: 10.4293/jsls.2020.00017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background and Objectives: Laparoscopy is the preferred method when operating in the abdomen. In this study, we evaluated systemic and morphological peritoneal cytokine modifications (RANTES/CCL5 and MCP-1/CCL2) due to CO2 pneumoperitoneum in rats. Methods: Twenty-five prepubertal Sprague-Dawley rats were randomized into three groups. Pneumoperitoneum lasting 30 minutes, was induced with a flow of 0.5 L/min, in two groups (S1 and S2, n = 20), at a P/CO2 of 6 and 10 mm Hg, respectively. In the control group (C, n = 5), only anesthesia was carried out. All animals were sacrificed after 24 hours. The serum of the rats was collected for ELISA, and the levels of the cytokines RANTES and MCP-1 were investigated. An immunohistochemical analysis of RANTES and MCP-1 was performed on samples of the peritoneum, and the morphological evaluation was conducted with a blinded evaluation by two independent, experienced pathologists by using a grading system (0, 1+, 2+, 3+: no, faint, moderate, and strong reactivity, respectively). Results: RANTES mean levels were significantly different in the S1, S2, and C groups (70.3 ± 2.26, 58.23 ± 4.32, 29.66 ± 4.03, respectively, P = .0001). The levels of MCP-1 were 32.1 ± 1.63 in the S1 group, 27.0 ± 9.26 in the S2 group, and 16.4 ± 9.55 in the C group (P = .159). Normal control peritoneum showed little reactivity, whereas a moderate to strong cytoplasmic reaction to anti-CCL5/CCL2 antibodies was observed in mesothelial and inflammatory cells in the S1 and S2 groups. Conclusion: CO2 pneumoperitoneum evokes an inflammatory response by modifying plasma RANTES levels and peritoneal CCL5/CCL2 expression.
Collapse
Affiliation(s)
- Alfonso Papparella
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | | | - Sara Ranucci
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Orlando Paciello
- Faculty of Veterinary Medicine, Department of Pathology and Animal Health, University of Naples Federico II, Naples, Italy
| | - Serenella Papparella
- Faculty of Veterinary Medicine, Department of Pathology and Animal Health, University of Naples Federico II, Naples, Italy
| | - Davide De Biase
- Faculty of Veterinary Medicine, Department of Pathology and Animal Health, University of Naples Federico II, Naples, Italy
| | - Grazia Cirillo
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Giuseppina Rosaria Umano
- Department of Woman, Child, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
76
|
Yan D, Liu X, Xu H, Guo SW. Mesothelial Cells Participate in Endometriosis Fibrogenesis Through Platelet-Induced Mesothelial-Mesenchymal Transition. J Clin Endocrinol Metab 2020; 105:5894452. [PMID: 32813013 DOI: 10.1210/clinem/dgaa550] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT While fibrosis in endometriosis has recently loomed prominently, the sources of myofibroblasts, the principal effector cell in fibrotic diseases, remain largely obscure. Mesothelial cells (MCs) can be converted into myofibroblasts through mesothelial-mesenchymal transition (MMT) in many fibrotic diseases and adhesion. OBJECTIVE To evaluate whether MCs contribute to the progression and fibrogenesis in endometriosis through MMT. SETTING, DESIGN, PATIENTS, INTERVENTION, AND MAIN OUTCOME MEASURES Dual immunofluorescence staining and immunohistochemistry using antibodies against calretinin, Wilms' tumor-1 (WT-1), and α-smooth muscle actin (α-SMA) were performed on lesion samples from 30 patients each with ovarian endometrioma (OE) and deep endometriosis (DE), and 30 normal endometrial (NE) tissue samples. Human pleural and peritoneal MCs were co-cultured with activated platelets or control medium with and without neutralization of transforming growth factor β1 (TGF-β1) and/or platelet-derived growth factor receptor (PDGFR) and their morphology, proliferation, and expression levels of genes and proteins known to be involved in MMT were evaluated, along with their migratory and invasive propensity, contractility, and collagen production. RESULTS The number of calretinin/WT-1 and α-SMA dual-positive fibroblasts in OE/DE lesions was significantly higher than NE samples. The extent of lesional fibrosis correlated positively with the lesional α-SMA staining levels. Human MCs co-cultured with activated platelets acquire a morphology suggestive of MMT, concomitant with increased proliferation, loss of calretinin expression, and marked increase in expression of mesenchymal markers. These changes coincided with functional differentiation as reflected by increased migratory and invasive capacity, contractility, and collagen production. Neutralization of TGF-β1 and PDGFR signaling abolished platelet-induced MMT in MCs. CONCLUSIONS MCs contribute to lesional progression and fibrosis through platelet-induced MMT.
Collapse
Affiliation(s)
- Dingmin Yan
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Hong Xu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
77
|
Wagner WL, Zheng Y, Pierce A, Ackermann M, Horstmann H, Kuner T, Ronchi P, Schwab Y, Konietzke P, Wünnemann F, Wielpütz MO, Kauczor HU, Mentzer SJ. Mesopolysaccharides: The extracellular surface layer of visceral organs. PLoS One 2020; 15:e0238798. [PMID: 32941441 PMCID: PMC7498049 DOI: 10.1371/journal.pone.0238798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 08/24/2020] [Indexed: 11/18/2022] Open
Abstract
The mesothelium is a dynamic and specialized tissue layer that covers the somatic cavities (pleural, peritoneal, and pericardial) as well as the surface of the visceral organs such as the lung, heart, liver, bowel and tunica vaginalis testis. The potential therapeutic manipulation of visceral organs has been complicated by the carbohydrate surface layer-here, called the mesopolysaccharide (MPS)-that coats the outer layer of the mesothelium. The traditional understanding of MPS structure has relied upon fixation techniques known to degrade carbohydrates. The recent development of carbohydrate-preserving fixation for high resolution imaging techniques has provided an opportunity to re-examine the structure of both the MPS and the visceral mesothelium. In this report, we used high pressure freezing (HPF) as well as serial section transmission electron microscopy to redefine the structure of the MPS expressed on the murine lung, heart and liver surface. Tissue preserved by HPF and examined by transmission electron microscopy demonstrated a pleural MPS layer 13.01±1.1 um deep-a 100-fold increase in depth compared to previously reported data obtained with conventional fixation techniques. At the base of the MPS were microvilli 1.1±0.35 um long and 42±5 nm in diameter. Morphological evidence suggested that the MPS was anchored to the mesothelium by microvilli. In addition, membrane pits 97±17 nm in diameter were observed in the apical mesothelial membrane. The spatial proximity and surface density (29±4.5%) of the pits suggested an active process linked to the structural maintenance of the MPS. The striking magnitude and complex structure of the MPS indicates that it is an important consideration in studies of the visceral mesothelium.
Collapse
Affiliation(s)
- Willi L. Wagner
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA, United States of America
| | - Yifan Zheng
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA, United States of America
| | - Aidan Pierce
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA, United States of America
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Heinz Horstmann
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, University of Heidelberg, Germany
| | - Thomas Kuner
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, University of Heidelberg, Germany
| | - Paolo Ronchi
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory, Electron Microscopy Core Facility, Heidelberg, Germany
| | - Yannick Schwab
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory, Electron Microscopy Core Facility, Heidelberg, Germany
- European Molecular Biology Laboratory, Cell Biology and Biophysics Unit, Heidelberg, Germany
| | - Philip Konietzke
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
| | - Felix Wünnemann
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
| | - Mark O. Wielpütz
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center, Member of the German Center for Lung Research, University of Heidelberg, Heidelberg, Germany
| | - Steven J. Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA, United States of America
| |
Collapse
|
78
|
Clements D, Miller S, Johnson SR. Pulmonary Lymphangioleiomyomatosis originates in the pleural mesothelial cell population. Med Hypotheses 2020; 141:109703. [PMID: 32276237 DOI: 10.1016/j.mehy.2020.109703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a cystic lung disease mainly affecting women, in which degradation of the lung parenchyma is associated with a cell of unknown provenance, known as a LAM cell. LAM cells carry TSC2 mutations and can be identified in the lung parenchyma by their expression of both smooth muscle actin and antigens characteristic of melanocytes and melanocytic tumors. The nature of the cell-of-origin of LAM is controversial, and despite continued research effort remains elusive. Further, it has not been possible to culture pulmonary LAM cells in vitro, and current research relies on cells and animal models which may not recapitulate all features of the disease. We noted aberrant expression of melanoma antigens in pleural mesothelial cells in lung tissue from LAM patients, indicating that these cells could be the precursors of parenchymal LAM cells. We hypothesise that loss of tuberin function following TSC2 mutation in the mesothelial cell lineage gives rise to the cell-of-origin of pulmonary LAM (P-LAM), and of other associated conditions commonly noted in LAM patients. The unique properties of mesothelial cells provide a straightforward explanation of the diverse presentation of LAM.
Collapse
Affiliation(s)
- D Clements
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK.
| | - S Miller
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK
| | - S R Johnson
- Division of Respiratory Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK; National Centre for Lymphangioleiomyomatosis, Nottingham University Hospitals NHS Trust, Queen's Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
79
|
The Selective Angiotensin II Type 2 Receptor Agonist Compound 21 Reduces Abdominal Adhesions in Mice. J Surg Res 2020; 256:231-242. [PMID: 32711180 DOI: 10.1016/j.jss.2020.06.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/28/2020] [Accepted: 06/16/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Abdominal adhesions (AAs) are post-traumatic fibrous bands that connect visceral and/or peritoneal surfaces, leading to possible long-term complications. The effect of a novel antifibrotic selective angiotensin II type 2 receptor agonist, compound 21 (C21) on AA formation was assessed in a murine model. METHODS Female BALB/c mice were laparotomized and the cecum and overlying parietal peritoneum abraded. C21 (10 μg/kg) or saline (vehicle) were administered orally or intraperitoneally daily. Mice were sacrificed 8 days after surgery, adhesions graded, and peritoneal fluid collected for transforming growth factor (TGF)-β levels. Laparotomy incisions were excised for immunohistochemistry. In vitro, scratch assays were performed using primary parietal peritoneal fibroblasts and visceral mesothelial cells treated with C21 (10 μM), angiotensin II (1 μM), or both. Western blot analysis of primary cell lysates was performed for total and phosphorylated SMAD 2/3. RESULTS Oral and intraperitoneal C21 reduced AA formation and TGF-β levels in peritoneal fluid. Surgical incisions demonstrated decreased α-smooth muscle actin expression in C21-treated animals, but no difference in vascularity, macrophage infiltration, collagen I/III distribution and density, and dermal thickness. Migration and expression of phosphorylated SMAD 2/3 was reduced in parietal peritoneal fibroblasts and visceral mesothelial cells treated with C21. CONCLUSIONS Local and systemic C21 administration reduced or completely prevented AA formation. These findings may be attributed to decreased intraperitoneal TGF-β in vivo and decreased migration of peritoneal fibroblasts and visceral mesothelial cells. Importantly, C21 did not have histologically quantifiable effects on laparotomy wounds, suggesting C21 could reduce AA formation without compromising laparotomy healing.
Collapse
|
80
|
Deng Z, Fear MW, Suk Choi Y, Wood FM, Allahham A, Mutsaers SE, Prêle CM. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int J Biochem Cell Biol 2020; 126:105802. [PMID: 32668329 DOI: 10.1016/j.biocel.2020.105802] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is characterised by excessive scarring in the lung which leads to compromised lung function, serious breathing problems and in some diseases, death. It includes several lung disorders with idiopathic pulmonary fibrosis (IPF) the most common and most severe. Pulmonary fibrosis is considered to be perpetuated by aberrant wound healing which leads to fibroblast accumulation, differentiation and activation, and deposition of excessive amounts of extracellular matrix (ECM) components, in particular, collagen. Recent studies have identified the importance of changes in the composition and structure of lung ECM during the development of pulmonary fibrosis and the interaction between ECM and lung cells. There is strong evidence that increased matrix stiffness induces changes in cell function including proliferation, migration, differentiation and activation. Understanding how changes in the ECM microenvironment influence cell behaviour during fibrogenesis, and the mechanisms regulating these changes, will provide insight for developing new treatments.
Collapse
Affiliation(s)
- Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital, Nedlands, WA, Australia; Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Amira Allahham
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
81
|
Si M, Wang Q, Li Y, Lin H, Luo D, Zhao W, Dou X, Liu J, Zhang H, Huang Y, Lou T, Hu Z, Peng H. Inhibition of hyperglycolysis in mesothelial cells prevents peritoneal fibrosis. Sci Transl Med 2020; 11:11/495/eaav5341. [PMID: 31167927 DOI: 10.1126/scitranslmed.aav5341] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Progressive peritoneal fibrosis affects patients receiving peritoneal dialysis (PD) and has no reliable treatment. The mechanisms that initiate and sustain peritoneal fibrosis remain incompletely elucidated. To overcome these problems, we developed a strategy that prevents peritoneal fibrosis by suppressing PD-stimulated mesothelial-to-mesenchymal transition (MMT). We evaluated single-cell transcriptomes of mesothelial cells obtained from normal peritoneal biopsy and effluent from PD-treated patients. In cells undergoing MMT, we found cellular heterogeneity and intermediate transition states associated with up-regulation of enzymes involved in glycolysis. The expression of glycolytic enzymes was correlated with the development of MMT. Using gene expression profiling and metabolomics analyses, we confirmed that PD fluid induces metabolic reprogramming, characterized as hyperglycolysis, in mouse peritoneum. We found that transforming growth factor β1 (TGF-β1) can substitute for PD fluid to stimulate hyperglycolysis, suppressing mitochondrial respiration in mesothelial cells. Blockade of hyperglycolysis with 2-deoxyglucose (2-DG) inhibited TGF-β1-induced profibrotic cellular phenotype and peritoneal fibrosis in mice. We developed a triad of adeno-associated viruses that overexpressed microRNA-26a and microRNA-200a while inhibiting microRNA-21a to target hyperglycolysis and fibrotic signaling. Intraperitoneal injection of the viral triad inhibited the development of peritoneal fibrosis induced by PD fluid in mice. We conclude that hyperglycolysis is responsible for MMT and peritoneal fibrogenesis, and this aberrant metabolic state can be corrected by modulating microRNAs in the peritoneum. These results could provide a therapeutic strategy to combat peritoneal fibrosis.
Collapse
Affiliation(s)
- Meijun Si
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qianqian Wang
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.,Nephrology Division, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
| | - Yin Li
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Hongchun Lin
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Dan Luo
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Wenbo Zhao
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xianrui Dou
- Nephrology Division, Shunde Hospital of Southern Medical University, Foshan 528300, China
| | - Jun Liu
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Zhang
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yong Huang
- Division of Gastrointestinal Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Tanqi Lou
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hui Peng
- Nephrology Division, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
82
|
Ojasalu K, Brehm C, Hartung K, Nischak M, Finkernagel F, Rexin P, Nist A, Pavlakis E, Stiewe T, Jansen JM, Wagner U, Gattenlöhner S, Bräuninger A, Müller-Brüsselbach S, Reinartz S, Müller R. Upregulation of mesothelial genes in ovarian carcinoma cells is associated with an unfavorable clinical outcome and the promotion of cancer cell adhesion. Mol Oncol 2020; 14:2142-2162. [PMID: 32533757 PMCID: PMC7463315 DOI: 10.1002/1878-0261.12749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/19/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
A hallmark of ovarian high‐grade serous carcinoma (HGSC) is its early and massive peritoneal dissemination via the peritoneal fluid. It is generally believed that tumor cells must breach the mesothelium of peritoneal organs to adhere to the underlying extracellular matrix (ECM) and initiate metastatic growth. However, the molecular mechanisms underlying these processes are only partially understood. Here, we have analyzed 52 matched samples of spheroids and solid tumor masses (suspected primary lesions and metastases) from 10 patients by targeted sequencing of 21 loci previously proposed as targets of HGSC driver mutations. This analysis revealed very similar patterns of genetic alterations in all samples. One exception was FAT3 with a strong enrichment of mutations in metastases compared with presumed primary lesions in two cases. FAT3 is a putative tumor suppressor gene that codes for an atypical cadherin, pointing a potential role in peritoneal dissemination in a subgroup of HGSC patients. By contrast, transcriptome data revealed clear and consistent differences between tumor cell spheroids from ascites and metastatic lesions, which were mirrored by the in vitro adherence of ascites‐derived spheroids. The adhesion‐induced transcriptional alterations in metastases and adherent cells resembled epithelial–mesenchymal transition, but surprisingly also included the upregulation of a specific subset of mesothelial genes, such as calretinin (CALB2) and podoplanin (PDPN). Consistent with this finding, calretinin staining was also observed in subsets of tumor cells in HGSC metastases, particularly at the invasive tumor edges. Intriguingly, a high expression of either CALB2 or PDPN was strongly associated with a poor clinical outcome. siRNA‐mediated CALB2 silencing triggered the detachment of adherent HGSC cells in vitro and inhibited the adhesion of detached HGSC cells to collagen type I. Our data suggest that the acquisition of a mesenchymal–mesothelial phenotype contributes to cancer cell adhesion to the ECM of peritoneal organs and HGSC progression.
Collapse
Affiliation(s)
- Kaire Ojasalu
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Corinna Brehm
- Institute of Pathology, Philipps University, Marburg, Germany
| | - Kristin Hartung
- Institute of Pathology, Justus-Liebig University, Giessen, Germany
| | - Maximilian Nischak
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Peter Rexin
- Institute of Pathology, Philipps University, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Evangelos Pavlakis
- Institute of Molecular Oncology, Member of the German Center of Lung Research (DZL), Philipps University, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, Marburg, Germany.,Institute of Molecular Oncology, Member of the German Center of Lung Research (DZL), Philipps University, Marburg, Germany
| | - Julia M Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital Giessen and Marburg (UKGM), Marburg, Germany
| | | | | | | | - Silke Reinartz
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Rolf Müller
- Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| |
Collapse
|
83
|
Yan D, Liu X, Guo SW. The establishment of a mouse model of deep endometriosis. Hum Reprod 2020; 34:235-247. [PMID: 30561644 DOI: 10.1093/humrep/dey361] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/30/2018] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Is it possible to establish a mouse model of deep endometriosis (DE)? SUMMARY ANSWER A mouse DE model that is macroscopically and microscopically similar to nodular lesions in humans can be constructed in as short as 3 weeks by intraperitoneal injection of uterine fragments along with the infusion of substance P (SP) and/or calcitonin gene-related peptide (CGRP). WHAT IS KNOWN ALREADY Although a baboon DE model was reported 5 years ago, its prohibitive cost and demand for facilities and expertise associated with the use of non-human primates put its use out of reach for most laboratories. STUDY DESIGN, SIZE, DURATION A total of 48 female Balb/C mice were used for this study. Among them, 16 were randomly selected as donors that contributed uterine fragments, and the remaining 32 were recipient mice. The mice with induced endometriosis were followed up for 3-4 weeks. PARTICIPANTS/MATERIALS, SETTING, METHODS One day before the induction of endometriosis by intraperitoneal injection of uterine fragments, osmotic pumps were inserted into equal groups of recipient mice to infuse either sterile saline, SP, CGRP, or both SP and CGRP. The hotplate test was administrated to all mice at the baseline and before and after induction of endometriosis. Four (3 for the SP+CGRP group) weeks after induction, all mice were sacrificed. Their endometriotic lesions were excised, weighed and processed for histopathologic examination, and histochemistry, immunohistochemistry and immunofluorescence analyses of markers of proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT), smooth muscle metaplasia (SMM), mesothelial-mesenchymal transition (MMT) and endothelial-mesenchymal transition (EndoMT) were done. The extent of lesional fibrosis was evaluated by Masson trichrome staining. To further evaluate surrounding organ/tissue invasion, the peritoneal areas adhesive to the lesions were excised for immunohistochemical analysis. MAIN RESULTS AND THE ROLE OF CHANCE Endometriotic lesions in mice treated with SP and/or CGRP satisfied all requirements for DE, i.e. presence of endometrial epithelial and stromal cells, abundance of fibromuscular content, and encapsulation in surrounding tissues or organs. The lesion weight in the CGRP, SP and SP+CGRP groups was 1.62, 2.14 and 2.18-fold, respectively, heavier than that of control group. Concomitantly, the SP, CGRP and SP+CGRP groups had significantly shorter hotplate latency than that of control group. Lesions in mice treated with SP and/or CGRP, especially with SP+CGRP, exhibited characteristics consistent with EMT, FMT, SMM and extensive fibrosis, along with signs of MMT and EndoMT. Lesional invasion into surrounding tissues/organs was found to be 25.0, 75.0 and 87.5% in mice treated with CGRP, SP and SP+CGRP, but none in control mice. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study is limited by the use of histologic and immunohistochemistry analyses only and lacks molecular data. WIDER IMPLICATIONS OF THE FINDINGS The establishment of a mouse DE model supports the idea that endometriotic lesions are wounds undergoing repeated tissue injury and repair and underscores the importance of microenvironments in shaping the lesions' destiny. In addition, signs consistent with MMT and EndoMT indicate that there may be more culpable factors that still remain unidentified and should be pursued in the future. Moreover, the close correlation between the extent of lesional fibrosis and markers of EMT, MMT, EndoMT, FMT and SMM as shown here should facilitate our understanding of the molecular mechanisms underlying the DE pathophysiology. Since this DE model is based on a biologically plausible and evidence-backed theory, it should shed much needed insight into the molecular mechanisms underlying the pathophysiology of DE. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by Grants 81471434 (S.W.G.), 81530040 (S.W.G.), 81771553 (S.W.G.), 81671436 (X.S.L.) and 81871144 (X.S.L.) from the National Natural Science Foundation of China. None of the authors has any conflict of interest to disclose.
Collapse
Affiliation(s)
- Dingmin Yan
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
84
|
Adipose-Derived Mesenchymal Stem Cells Modulate Fibrosis and Inflammation in the Peritoneal Fibrosis Model Developed in Uremic Rats. Stem Cells Int 2020; 2020:3768718. [PMID: 32565826 PMCID: PMC7256710 DOI: 10.1155/2020/3768718] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/17/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
Peritoneal fibrosis (PF) represents a long-term complication of peritoneal dialysis (PD), affecting the peritoneal membrane (PM) function. Adipose tissue-derived mesenchymal stem cells (ASC) display immunomodulatory effects and may represent a strategy to block PF. The aim of this study was to analyze the effect of ASC in an experimental PF model developed in uremic rats. To mimic the clinical situation of patients on long-term PD, a combo model, characterized by the combination of PF and chronic kidney disease (CKD), was developed in Wistar rats. Rats were fed with a 0.75% adenine-containing diet, for 30 days, to induce CKD with uremia. PF was induced with intraperitoneal injections of chlorhexidine gluconate (CG) from day 15 to 30. 1 × 106 ASC were intravenously injected at days 15 and 21. Rats were divided into 5 groups: control, normal rats; CKD, rats receiving adenine diet; PF, rats receiving CG; CKD+PF, CKD rats with PF; CKD+PF+ASC, uremic rats with PF treated with ASC. PF was assessed by Masson trichrome staining. Inflammation- and fibrosis-associated factors were assessed by immunohistochemistry, multiplex analysis, and qPCR. When compared with the control and CKD groups, GC administration induced a striking increase in PM thickness and inflammation in the PF and CKD+PF groups. The development of PF was blocked by ASC treatment. Further, the upregulation of profibrotic factors (TGF-β, fibronectin, and collagen) and the increased myofibroblast expression observed in the CKD+PF group were significantly ameliorated by ASC. Beyond the antifibrotic effect, ASC showed an anti-inflammatory effect avoiding leucocyte infiltration and the overexpression of inflammatory cytokines (IL-1β, TNF-α, and IL-6) in the PM induced by GC. ASC were effective in preventing the development of PF in the experimental model of CKD+PF, probably due to their immunomodulatory properties. These results suggest that ASC may represent a potential strategy for treating long-term PD-associated fibrosis.
Collapse
|
85
|
Abbott DM, Bortolotto C, Benvenuti S, Lancia A, Filippi AR, Stella GM. Malignant Pleural Mesothelioma: Genetic and Microenviromental Heterogeneity as an Unexpected Reading Frame and Therapeutic Challenge. Cancers (Basel) 2020; 12:cancers12051186. [PMID: 32392897 PMCID: PMC7281319 DOI: 10.3390/cancers12051186] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
Mesothelioma is a malignancy of serosal membranes including the peritoneum, pleura, pericardium and the tunica vaginalis of the testes. Malignant mesothelioma (MM) is a rare disease with a global incidence in countries like Italy of about 1.15 per 100,000 inhabitants. Malignant Pleural Mesothelioma (MPM) is the most common form of mesothelioma, accounting for approximately 80% of disease. Although rare in the global population, mesothelioma is linked to industrial pollutants and mineral fiber exposure, with approximately 80% of cases linked to asbestos. Due to the persistent asbestos exposure in many countries, a worldwide progressive increase in MPM incidence is expected for the current and coming years. The tumor grows in a loco-regional pattern, spreading from the parietal to the visceral pleura and invading the surrounding structures that induce the clinical picture of pleural effusion, pain and dyspnea. Distant spreading and metastasis are rarely observed, and most patients die from the burden of the primary tumor. Currently, there are no effective treatments for MPM, and the prognosis is invariably poor. Some studies average the prognosis to be roughly one-year after diagnosis. The uniquely poor mutational landscape which characterizes MPM appears to derive from a selective pressure operated by the environment; thus, inflammation and immune response emerge as key players in driving MPM progression and represent promising therapeutic targets. Here we recapitulate current knowledge on MPM with focus on the emerging network between genetic asset and inflammatory microenvironment which characterize the disease as amenable target for novel therapeutic approaches.
Collapse
Affiliation(s)
- David Michael Abbott
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy;
| | - Chandra Bortolotto
- Unit of Radiology, Department of Intensive Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy;
| | - Silvia Benvenuti
- Candiolo Cancer Institute, FPO—IRCCS—Str. Prov.le 142, km. 3,95—10060 Candiolo (TO), Italy;
| | - Andrea Lancia
- Unit of Radiation Therapy, Department of Medical Sciences and Infective Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy; (A.L.); (A.R.F.)
| | - Andrea Riccardo Filippi
- Unit of Radiation Therapy, Department of Medical Sciences and Infective Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy; (A.L.); (A.R.F.)
| | - Giulia Maria Stella
- Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, 27100 Pavia, Italy;
- Correspondence:
| |
Collapse
|
86
|
Gordillo CH, Sandoval P, Muñoz-Hernández P, Pascual-Antón L, López-Cabrera M, Jiménez-Heffernan JA. Mesothelial-to-Mesenchymal Transition Contributes to the Generation of Carcinoma-Associated Fibroblasts in Locally Advanced Primary Colorectal Carcinomas. Cancers (Basel) 2020; 12:cancers12020499. [PMID: 32098058 PMCID: PMC7072259 DOI: 10.3390/cancers12020499] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
During peritoneal metastasis, cancer cells spread from abdominal solid tumors, disseminate through the peritoneal fluid and attach to and invade through mesothelial cells (MCs) that line the peritoneum. Intestinal adenocarcinomas originating in the mucosa infiltrate the submucosa, muscle layer, and serosa in order to finally colonize the peritoneal cavity. However, the mechanism by which metastatic cells leave the primary tumor and reach the peritoneal cavity has not been previously described. Hence, we investigate whether MCs lining visceral peritoneum, through a mesothelial-to-mesenchymal transition (MMT), are a source of carcinoma-associated fibroblasts (CAFs), which could contribute to cancer progression toward the peritoneal cavity. CAFs detected in biopsies from patients with superficially invasive colorectal cancer differed from locally advanced tumors. An aberrant accumulation of myofibroblasts expressing mesothelial markers was found in the stroma of deeply infiltrative tumors located in the neighborhood of a frequently activated mesothelium. We suggest that MMT is a key event in the early stages of peritoneal dissemination.
Collapse
Affiliation(s)
- Carlos H. Gordillo
- Servicio de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain (P.M.-H.)
| | - Pilar Sandoval
- Centro de Biología Molecular “Severo Ochoa”—CSIC, 28049 Madrid, Spain; (P.S.); (L.P.-A.)
| | - Patricia Muñoz-Hernández
- Servicio de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain (P.M.-H.)
| | - Lucía Pascual-Antón
- Centro de Biología Molecular “Severo Ochoa”—CSIC, 28049 Madrid, Spain; (P.S.); (L.P.-A.)
| | - Manuel López-Cabrera
- Centro de Biología Molecular “Severo Ochoa”—CSIC, 28049 Madrid, Spain; (P.S.); (L.P.-A.)
- Correspondence: (M.L.-C.); (J.A.J.-H.)
| | - José A. Jiménez-Heffernan
- Servicio de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain (P.M.-H.)
- Correspondence: (M.L.-C.); (J.A.J.-H.)
| |
Collapse
|
87
|
Laukka M, Hoppela E, Salo J, Rantakari P, Gronroos TJ, Orte K, Auvinen K, Salmi M, Gerke H, Thol K, Peuhu E, Kauhanen S, Merilahti P, Hartiala P. Preperitoneal Fat Grafting Inhibits the Formation of Intra-abdominal Adhesions in Mice. J Gastrointest Surg 2020; 24:2838-2848. [PMID: 31823326 PMCID: PMC7674570 DOI: 10.1007/s11605-019-04425-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/25/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Adhesion formation contributes to postoperative complications in abdominal and gynaecological surgery. Thus far, the prevention and treatment strategies have focused on mechanical barriers in solid and liquid form, but these methods are not in routine use. As autologous fat grafting has become popular in treatment of hypertrophic scars because of its immunomodulatory effects, we postulated that fat grafting could also prevent peritoneal adhesion through similar mechanisms. METHODS This was a control versus intervention study to evaluate the effect of fat grafting in the prevention on peritoneal adhesion formation. An experimental mouse model for moderate and extensive peritoneal adhesions was used (n = 4-6 mice/group). Adhesions were induced mechanically, and a free epididymal fat graft from wild type or CAG-DsRed mice was injected preperitoneally immediately after adhesion induction. PET/CT imaging and scaling of the adhesions were performed, and samples were taken for further analysis at 7 and 30 days postoperation. Macrophage phenotyping was further performed from peritoneal lavage samples, and the expression of inflammatory cytokines and mesothelial layer recovery were analysed from peritoneal tissue samples. RESULTS Fat grafting significantly inhibited the formation of adhesions. PET/CT results did not show prolonged inflammation in any of the groups. While the expression of anti-inflammatory and anti-fibrotic IL-10 was significantly increased in the peritoneum of the fat graft-treated group at 7 days, tissue-resident and repairing M2 macrophages could no longer be detected in the fat graft at this time point. The percentage of the continuous, healed peritoneum as shown by Keratin 8 staining was greater in the fat graft-treated group after 7 days. CONCLUSIONS Fat grafting can inhibit the formation of peritoneal adhesions in mice. Our results suggest that fat grafting promotes the peritoneal healing process in a paracrine manner thereby enabling rapid regeneration of the peritoneal mesothelial cell layer.
Collapse
Affiliation(s)
- Mervi Laukka
- Department of Plastic and General Surgery, Turku University Hospital, Turku, Finland ,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Erika Hoppela
- Department of Plastic and General Surgery, Turku University Hospital, Turku, Finland
| | - Jemiina Salo
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pia Rantakari
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tove J. Gronroos
- Turku PET Centre, University of Turku, Turku, Finland ,Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Katri Orte
- Genetics and Saske Tyks Laboratory Division, Turku University Hospital, Turku, Finland
| | - Kaisa Auvinen
- Institute of Biomedicine, University of Turku, Turku, Finland ,Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Marko Salmi
- Institute of Biomedicine, University of Turku, Turku, Finland ,Medicity Research Laboratories, University of Turku, Turku, Finland
| | - Heidi Gerke
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kerstin Thol
- Centre for Biotechnology, University of Turku, Turku, Finland
| | - Emilia Peuhu
- Centre for Biotechnology, University of Turku, Turku, Finland ,FICAN West Cancer Laboratory, Turku University Hospital and University of Turku, Turku, Finland
| | - Saila Kauhanen
- Department of Gastrointestinal Surgery, Turku University Hospital, Turku, Finland
| | - Pirjo Merilahti
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pauliina Hartiala
- Department of Plastic and General Surgery, Turku University Hospital, Turku, Finland ,Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
88
|
Tong JWV, Lingam P, Shelat VG. Adhesive small bowel obstruction - an update. Acute Med Surg 2020; 7:e587. [PMID: 33173587 PMCID: PMC7642618 DOI: 10.1002/ams2.587] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Small bowel obstruction (SBO) accounts for 12-16% of emergency surgical admissions and 20% of emergency surgical procedures. Even with the advent of laparoscopic surgery, intra-abdominal adhesions remain a significant cause of SBO, accounting for 65% of cases. History and physical examination are essential to identify signs of bowel ischemia as this indicates a need for urgent surgical exploration. Another critical aspect of evaluation includes establishing the underlying cause for obstruction and distinguishing between adhesive and non-adhesive etiologies as adhesive SBO (ASBO) can be managed non-operatively in 70-90% of patients. A patient with a history of abdominopelvic surgery along with one or more cardinal features of obstruction should be suspected to have ASBO until proven otherwise. Triad of severe pain, pain out of proportion to the clinical findings, and presence of an abdominal scar suggest possible closed-loop obstruction. Computed tomography has higher sensitivity and specificity compared to plain films and is recommended by the Bologna guidelines. Correcting fluid and electrolyte imbalance is an initial crucial step to mitigate severe hypovolemia. Patients should proceed with surgery if symptoms of bowel compromise are present, or if symptoms do not resolve or have worsened. Surgery is indicated in patients with ischemia, strangulation, perforation, peritonitis, or failure of non-operative treatment. With advances in minimal access technology and increasing experience, laparoscopic adhesiolysis is recommended. Mechanical adhesion barriers are an effective measure to prevent adhesion formation.
Collapse
Affiliation(s)
- Jia Wei Valerie Tong
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Pravin Lingam
- Department of General SurgeryTan Tock Seng HospitalSingaporeSingapore
| | | |
Collapse
|
89
|
Urso L, Cavallari I, Sharova E, Ciccarese F, Pasello G, Ciminale V. Metabolic rewiring and redox alterations in malignant pleural mesothelioma. Br J Cancer 2020; 122:52-61. [PMID: 31819191 PMCID: PMC6964675 DOI: 10.1038/s41416-019-0661-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/21/2019] [Accepted: 11/04/2019] [Indexed: 02/08/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy of mesothelial cells with increasing incidence, and in many cases, dismal prognosis due to its aggressiveness and lack of effective therapies. Environmental and occupational exposure to asbestos is considered the main aetiological factor for MPM. Inhaled asbestos fibres accumulate in the lungs and induce the generation of reactive oxygen species (ROS) due to the presence of iron associated with the fibrous silicates and to the activation of macrophages and inflammation. Chronic inflammation and a ROS-enriched microenvironment can foster the malignant transformation of mesothelial cells. In addition, MPM cells have a highly glycolytic metabolic profile and are positive in 18F-FDG PET analysis. Loss-of-function mutations of BRCA-associated protein 1 (BAP1) are a major contributor to the metabolic rewiring of MPM cells. A subset of MPM tumours show loss of the methyladenosine phosphorylase (MTAP) locus, resulting in profound alterations in polyamine metabolism, ATP and methionine salvage pathways, as well as changes in epigenetic control of gene expression. This review provides an overview of the perturbations in metabolism and ROS homoeostasis of MPM cells and the role of these alterations in malignant transformation and tumour progression.
Collapse
Affiliation(s)
- Loredana Urso
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | | | | | | | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| |
Collapse
|
90
|
Muhar AM, Putra A, Warli SM, Munir D. Hypoxia-Mesenchymal Stem Cells Inhibit Intra-Peritoneal Adhesions Formation by Upregulation of the IL-10 Expression. Open Access Maced J Med Sci 2019; 7:3937-3943. [PMID: 32165932 PMCID: PMC7061407 DOI: 10.3889/oamjms.2019.713] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/19/2019] [Accepted: 10/20/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Intra-peritoneal adhesions (IPAs) common occurre in post abdominal surgical. Athough many methods have been developed for controlling IPAs, including mesenchymal stem cells (MSCs) application, however, there is none completely preventing in due to the mesothelial structure may promote the prolonged inflammations leading. Nevertheless hypoxia-MSCs (H-MSCs) have more potent in controlling the inflammation than normoxia-MSCs (N-MSCs) by releasing several anti-inflamation particularly IL-10, however the H-MSCs application to inhibit IPAs remain unclear. AIM The aim of this study was to investigate the effectiveness of H-MSCs in preventing the AIPs event by releasing IL-10 on the ileum abrasion sutured omental patch as the animal model of peritoneal adhesion. METHODS Using 24 IPAs animal model were randomly divided into 4 groups: Sham (Sh), Control (C), H-MSCs at high dose (T1) and H-MSCs at low dose (T2). H-MSCs were incubated under hypoxic conditions (5% O2), 37°C and 5% CO2 for 24 hours. The expression level of IL-10 was performed using RT-PCR analysis. The macroscopic appearance of IPAs was evaluated using Nair's scale base on the absence/presence of adhesion, whereas the microscopic by Zuhlke's scale at Hematoxylin and eosin (H&E) staining. RESULTS This study showed a significanly increase in IL-10 expression (p < 0.05) at all T groups. In line with this, we also found a significant difference in IPAs between T groups and Control as well as a Sham (p < 0.05) either in the macroscopic or microscopic analysis. CONCLUSION H-MSCs has a robust ability in inhibiting severe IPAs characterized by the decreased of adhesion formation and the enhanced expression of IL-10.
Collapse
Affiliation(s)
- Adi Muradi Muhar
- Department of Doctoral Degree Program, Medical Faculty, Universitas Sumatera Utara, Medan, North Sumatera, Indonesia
| | - Agung Putra
- Stem Cell And Cancer Research (SCCR), Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Central Java, Indonesia.,Department of Postgraduate Biomedical Science, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Central Java, Indonesia.,Department of Pathological Anatomy, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Central Java, Indonesia
| | - Syah Mirsya Warli
- Department of Urology, Medical Faculty, Universitas Sumatera Utara, Medan, North Sumatera, Indonesia
| | - Delfitri Munir
- Pusat Unggulan Inovasi (PUI) Stem Cell, Universitas Sumatera Utara (USU), Medan, North Sumatera, Indonesia
| |
Collapse
|
91
|
Jagirdar RM, Bozikas A, Zarogiannis SG, Bartosova M, Schmitt CP, Liakopoulos V. Encapsulating Peritoneal Sclerosis: Pathophysiology and Current Treatment Options. Int J Mol Sci 2019; 20:ijms20225765. [PMID: 31744097 PMCID: PMC6887950 DOI: 10.3390/ijms20225765] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022] Open
Abstract
Encapsulating peritoneal sclerosis (EPS) is a life-threatening complication of long-term peritoneal dialysis (PD), which may even occur after patients have switched to hemodialysis (HD) or undergone kidney transplantation. The incidence of EPS varies across the globe and increases with PD vintage. Causative factors are the chronic exposure to bioincompatible PD solutions, which cause long-term modifications of the peritoneum, a high peritoneal transporter status involving high glucose concentrations, peritonitis episodes, and smoldering peritoneal inflammation. Additional potential causes are predisposing genetic factors and some medications. Clinical symptoms comprise signs of intestinal obstruction and a high peritoneal transporter status with incipient ultrafiltration failure. In radiological, macro-, and microscopic studies, a massively fibrotic and calcified peritoneum enclosed the intestine and parietal wall in such cases. Empirical treatments commonly used are corticosteroids and tamoxifen, which has fibrinolytic properties. Immunosuppressants like azathioprine, mycophenolate mofetil, or mTOR inhibitors may also help with reducing inflammation, fibrin deposition, and collagen synthesis and maturation. In animal studies, N-acetylcysteine, colchicine, rosiglitazone, thalidomide, and renin-angiotensin system (RAS) inhibitors yielded promising results. Surgical treatment has mainly been performed in severe cases of intestinal obstruction, with varying results. Mortality rates are still 25–55% in adults and about 14% in children. To reduce the incidence of EPS and improve the outcome of this devastating complication of chronic PD, vigorous consideration of the risk factors, early diagnosis, and timely discontinuation of PD and therapeutic interventions are mandatory, even though these are merely based on empirical evidence.
Collapse
Affiliation(s)
- Rajesh M. Jagirdar
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (R.M.J.); (A.B.)
| | - Andreas Bozikas
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (R.M.J.); (A.B.)
| | - Sotirios G. Zarogiannis
- Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.G.Z.); (M.B.); (C.P.S.)
- Department of Physiology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.G.Z.); (M.B.); (C.P.S.)
| | - Claus Peter Schmitt
- Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany; (S.G.Z.); (M.B.); (C.P.S.)
| | - Vassilios Liakopoulos
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (R.M.J.); (A.B.)
- Correspondence: ; Tel.: +30-2310-994694
| |
Collapse
|
92
|
Birkbeck R, Humm K, Cortellini S. A review of hyperfibrinolysis in cats and dogs. J Small Anim Pract 2019; 60:641-655. [PMID: 31608455 DOI: 10.1111/jsap.13068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
The fibrinolytic system is activated concurrently with coagulation; it regulates haemostasis and prevents thrombosis by restricting clot formation to the area of vascular injury and dismantling the clot as healing occurs. Dysregulation of the fibrinolytic system, which results in hyperfibrinolysis, may manifest as clinically important haemorrhage. Hyperfibrinolysis occurs in cats and dogs secondary to a variety of congenital and acquired disorders. Acquired disorders associated with hyperfibrinolysis, such as trauma, cavitary effusions, liver disease and Angiostrongylus vasorum infection, are commonly encountered in primary care practice. In addition, delayed haemorrhage reported in greyhounds following trauma and routine surgical procedures has been attributed to a hyperfibrinolytic disorder, although this has yet to be characterised. The diagnosis of hyperfibrinolysis is challenging and, until recently, has relied on techniques that are not readily available outside referral hospitals. With the recent development of point-of-care viscoelastic techniques, assessment of fibrinolysis is now possible in referral practice. This will provide the opportunity to target haemorrhage due to hyperfibrinolysis with antifibrinolytic drugs and thereby reduce associated morbidity and mortality. The fibrinolytic system and the conditions associated with increased fibrinolytic activity in cats and dogs are the focus of this review article. In addition, laboratory and point-of-care techniques for assessing hyperfibrinolysis and antifibrinolytic treatment for patients with haemorrhage are reviewed.
Collapse
Affiliation(s)
- R Birkbeck
- Department of Veterinary Clinical Sciences, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| | - K Humm
- Department of Veterinary Clinical Sciences, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| | - S Cortellini
- Department of Veterinary Clinical Sciences, The Royal Veterinary College, Hertfordshire, AL9 7TA, UK
| |
Collapse
|
93
|
Wu J, Huang Q, Li P, Wang Y, Zheng C, Lei X, Li S, Gong W, Yin B, Luo C, Xiao J, Zhou W, Xu Z, Chen Y, Peng F, Long H. MicroRNA-145 promotes the epithelial-mesenchymal transition in peritoneal dialysis-associated fibrosis by suppressing fibroblast growth factor 10. J Biol Chem 2019; 294:15052-15067. [PMID: 31431501 PMCID: PMC6791318 DOI: 10.1074/jbc.ra119.007404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
Peritoneal fibrosis is a common complication of long-term peritoneal dialysis (PD) and the principal cause of ultrafiltration failure during PD. The initial and reversible step in PD-associated peritoneal fibrosis is the epithelial-mesenchymal transition (EMT). Although the mechanisms in the EMT have been the focus of many studies, only limited information is currently available concerning microRNA (miRNA) regulation in peritoneal fibrosis. In this study, we aimed to characterize the roles of microRNA-145 (miR-145) and fibroblast growth factor 10 (FGF10) in peritoneal fibrosis. After inducing EMT with transforming growth factor-β1 (TGF-β1) in vitro, we found that miR-145 is significantly up-regulated, whereas FGF10 is markedly down-regulated, suggesting a close link between miR-145 and FGF10 in peritoneal fibrosis, further confirmed in luciferase reporter experiments. Furthermore, in human peritoneal mesothelial cells (i.e. HMrSV5 cells), miR-145 mimics induced EMT, whereas miR-145 inhibition suppressed EMT, and we also observed that miR-145 suppressed FGF10 expression. In vivo, we found that the exogenous delivery of an miR-145 expression plasmid both blocked FGF10 and intensified the EMT, whereas miR-145 inhibition promoted the expression of FGF10 and reversed the EMT. In conclusion, miR-145 promotes the EMT during the development of peritoneal fibrosis by suppressing FGF10 activity, suggesting that miR-145 represents a potential therapeutic target for managing peritoneal fibrosis.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qianyin Huang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Peilin Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxian Wang
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chenghao Zheng
- Second Clinical Medical School, Southern Medical University, Guangzhou 510280, China
| | - Xianghong Lei
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wangqiu Gong
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bohui Yin
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weidong Zhou
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhaozhong Xu
- Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yihua Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
94
|
Moscoso CG, Steer CJ. "Let my liver rather heat with wine" - a review of hepatic fibrosis pathophysiology and emerging therapeutics. Hepat Med 2019; 11:109-129. [PMID: 31565001 PMCID: PMC6731525 DOI: 10.2147/hmer.s213397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/17/2019] [Indexed: 12/12/2022] Open
Abstract
Cirrhosis is characterized by extensive hepatic fibrosis, and it is the 14th leading cause of death worldwide. Numerous contributing conditions have been implicated in its development, including infectious etiologies, medication overdose or adverse effects, ingestible toxins, autoimmunity, hemochromatosis, Wilson’s disease and primary biliary cholangitis to list a few. It is associated with portal hypertension and its stigmata (varices, ascites, hepatic encephalopathy, combined coagulopathy and thrombophilia), and it is a major risk factor for hepatocellular carcinoma. Currently, orthotopic liver transplantation has been the only curative modality to treat cirrhosis, and the scarcity of donors results in many people waiting years for a transplant. Identification of novel targets for pharmacologic therapy through elucidation of key mechanistic components to induce fibrosis reversal is the subject of intense research. Development of robust models of hepatic fibrosis to faithfully characterize the interplay between activated hepatic stellate cells (the principal fibrogenic contributor to fibrosis initiation and perpetuation), hepatocytes and extracellular matrix components has the potential to identify critical components and mechanisms that can be exploited for targeted treatment. In this review, we will highlight key cellular pathways involved in the pathophysiology of fibrosis from extracellular ligands, effectors and receptors, to nuclear receptors, epigenetic mechanisms, energy homeostasis and cytokines. Further, molecular pathways of hepatic stellate cell deactivation are discussed, including apoptosis, senescence and reversal or transdifferentiation to an inactivated state resembling quiescence. Lastly, clinical evidence of fibrosis reversal induced by biologics and small molecules is summarized, current compounds under clinical trials are described and efforts for treatment of hepatic fibrosis with mesenchymal stem cells are highlighted. An enhanced understanding of the rich tapestry of cellular processes identified in the initiation, perpetuation and resolution of hepatic fibrosis, driven principally through phenotypic switching of hepatic stellate cells, should lead to a breakthrough in potential therapeutic modalities.
Collapse
Affiliation(s)
- Carlos G Moscoso
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition
| | - Clifford J Steer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition.,Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
95
|
Danhong Injection Alleviates Postoperative Intra-abdominal Adhesion in a Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4591384. [PMID: 31531183 PMCID: PMC6721271 DOI: 10.1155/2019/4591384] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
Background Among all the common complications that occur after abdominal surgery, intestinal adhesion is perhaps the most unpleasant one. However, current methods to treat and prevent intestinal adhesion are limited; thus, exploring new methods to prevent and treat intestinal adhesion is greatly needed. In this study, we demonstrated that Danhong injection (DHI) may be used as a promising method to prevent and treat intra-abdominal adhesion in a rat model. Materials and Methods Forty-eight rats were randomly divided into six groups. Except for the sham-operated group, all rats underwent cecal abrasion to establish an adhesion model. After the operation, the rats in the DHI-treated groups received different doses of DHI via the tail vein daily, while the other group was treated with the same volume of saline solution. Seven days after the operation, all rats were sacrificed, and the degree of adhesion was evaluated by Nair's scoring system. The extent of inflammation in the adhesion tissue was detected by HE staining and the expression of tumor necrosis factor-α (TNF-α) and transforming growth factor-β (TGF-β). The collagen deposition was assessed by Sirius red staining and α-SMA, MMP9, t-PA, and PAI-1 levels. Oxidative stress was indicated by the level of reactive oxygen species (ROS) in adhesion tissues and by immunohistochemical labeling of Nrf2. Furthermore, rat primary peritoneal mesothelial cells (RPMCs) were treated with H2O2 and DHI, and NF-κB phosphorylation was detected to illustrate the effect of DHI on oxidative stress. Results The intra-abdominal adhesion scores were significantly decreased in the groups treated with a high dose of DHI compared with the control groups, and the degree of inflammation, fibrosis, and oxidative stress was also significantly decreased. DHI treatment significantly reduced the levels of TNF-α, TGF-β1, and PAI and increased the expression levels of MMP9, Nrf2, and t-PA in the adhesion tissues. ROS levels and NF-κB phosphorylation were significantly reduced in DHI-treated RPMCs compared with the control RPMCs. Conclusion DHI alleviates the formation of postoperative intra-abdominal adhesions by inhibiting inflammation, collagen deposition, and oxidative stress in a rat model and may serve as a promising drug to prevent intra-abdominal adhesions.
Collapse
|
96
|
Qi P, Zheng YG, Ohta S, Kokudo N, Hasegawa K, Ito T. In Situ Fabrication of Double-Layered Hydrogels via Spray Processes to Prevent Postoperative Peritoneal Adhesion. ACS Biomater Sci Eng 2019; 5:4790-4798. [DOI: 10.1021/acsbiomaterials.9b00791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | | | - Norihiro Kokudo
- National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | | | | |
Collapse
|
97
|
Li Y, Xiao Z, Zhou Y, Zheng S, An Y, Huang W, He H, Yang Y, Li S, Chen Y, Xiao J, Wu J. Controlling the Multiscale Network Structure of Fibers To Stimulate Wound Matrix Rebuilding by Fibroblast Differentiation. ACS APPLIED MATERIALS & INTERFACES 2019; 11:28377-28386. [PMID: 31251577 DOI: 10.1021/acsami.9b06439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The extracellular matrix (ECM) plays the role of a double-edged sword for controlling the differentiation of fibroblasts toward contractile myofibroblasts in the wound healing process. However, the exact structure-function relationship between ECM morphology and fibroblast behaviors still remains unclear. To better understand this relationship, herein, we designed and prepared a series of biocompatible polycaprolactone (PCL)-based fibers with different fiber diameters (nano vs micro) and different alignments (random vs aligned) using a simple electrospinning process, with a particular attention to the morphological effect of PCL fiber scaffolds on guiding fibroblast behaviors. Microfibers with the larger fiber diameters induce less cell spreading, adhesion, differentiation, and migration because of their lower surface tension. In contrast, nanofibers will retain fibroblast cells with typical spindle shapes and promote the expression of focal adhesion proteins through the integrin pathway. Furthermore, nanofibers upregulate the expression of α-smooth muscle actin (α-SMA), transforming growth factor, and vimentin filaments, indicating that the size change of the PCL fiber matrix from micrometers to nanometers indeed alters fibroblast differentiation to activate more α-SMA-expressed contractile myofibroblasts. Such a fiber size-dependent fibroblast behavior is largely attributed to the enhanced surface tension from the dressing matrix, which helps to promote the conversion of fibroblasts to myofibroblasts via either tissue regeneration or fibrosis. Therefore, this work further indicated that the rearrangement of collagen from nano-tropocollagen to micro-collagen bundles during the wound healing process can reverse fibroblasts to myofibroblasts from motivated to demise. This finding allows us to achieve the structural-based design of a new fibrous matrix for promoting wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Yi Li
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
- The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325200 , P. R. China
| | - Zecong Xiao
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Yajiao Zhou
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Sen Zheng
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Ying An
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Wen Huang
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Huacheng He
- College of Chemistry and Materials Engineering , Wenzhou University , Wenzhou , Zhejiang 325027 , P. R. China
| | - Yao Yang
- College of Chemistry and Materials Engineering , Wenzhou University , Wenzhou , Zhejiang 325027 , P. R. China
| | - Shengyu Li
- College of Chemistry and Materials Engineering , Wenzhou University , Wenzhou , Zhejiang 325027 , P. R. China
| | - Yanxin Chen
- College of Chemistry and Materials Engineering , Wenzhou University , Wenzhou , Zhejiang 325027 , P. R. China
| | - Jian Xiao
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| | - Jiang Wu
- School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , P. R. China
| |
Collapse
|
98
|
High Proliferative Placenta-Derived Multipotent Cells Express Cytokeratin 7 at Low Level. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2098749. [PMID: 31392209 PMCID: PMC6662495 DOI: 10.1155/2019/2098749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 05/30/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to investigate the immunophenotypes and gene expression profile of high proliferative placenta-derived multipotent cells (PDMCs) population at different stages of culture. We demonstrated that the colonies resulting from single cells were either positive or negative for CK7, whereas only PDMC clones with weak CK7 expression (CK7low-clones) were highly proliferative. Interestingly, vimentin positive (Vim+) placental stromal mesenchymal cells did not express CK7 in situ, but double CK7+Vim+ cells detection in tissue explants and explants outgrowth indicated CK7 inducible expression in vitro. PCNA presence in CK7+Vim+ cells during placental explants culturing confirmed belonging of these cells to proliferative subpopulation. Transcription factors CDX2 and EOMES were expressed in both CK7low-clones and subset of stromal mesenchymal cells of first-trimester placental tissue in situ. Meanwhile, CK7low -clones and stromal mesenchymal cells of full-term placental tissue in situ expressed ERG heterogeneously. SPP1, COL2A1, and PPARG2 mesodermal-related genes expression by CK7low-clones additionally confirms their mesenchymal origin. Inherent stem cell-related gene expression (IFTM3, POU5F1, and VASA) in CK7low-clones might indicate their enrichment for progenitors. Finally, in CK7low-clones we observed expression of such trophoblast-associated genes as CGB types I and II, fusogenic ERVW-1, GCM1, and GATA3. Thus, our results indicate that PDMCs acquired the representative immunophenotype signature under culture conditions.
Collapse
|
99
|
Buechler MB, Kim KW, Onufer EJ, Williams JW, Little CC, Dominguez CX, Li Q, Sandoval W, Cooper JE, Harris CA, Junttila MR, Randolph GJ, Turley SJ. A Stromal Niche Defined by Expression of the Transcription Factor WT1 Mediates Programming and Homeostasis of Cavity-Resident Macrophages. Immunity 2019; 51:119-130.e5. [PMID: 31231034 DOI: 10.1016/j.immuni.2019.05.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/20/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022]
Abstract
Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Ki-Wook Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emily J Onufer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jesse W Williams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine C Little
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Claudia X Dominguez
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Qingling Li
- Microchemistry and Proteomics, Genentech, South San Francisco, CA 94080, USA
| | - Wendy Sandoval
- Microchemistry and Proteomics, Genentech, South San Francisco, CA 94080, USA
| | - Jonathan E Cooper
- Translational Oncology, Genentech, South San Francisco, CA 94080, USA
| | - Charles A Harris
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
100
|
Rouka E, Beltsios E, Goundaroulis D, Vavougios GD, Solenov EI, Hatzoglou C, Gourgoulianis KI, Zarogiannis SG. In Silico Transcriptomic Analysis of Wound-Healing-Associated Genes in Malignant Pleural Mesothelioma. ACTA ACUST UNITED AC 2019; 55:medicina55060267. [PMID: 31212858 PMCID: PMC6631992 DOI: 10.3390/medicina55060267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
Background and objectives: Malignant pleural mesothelioma (MPM) is a devastating malignancy with poor prognosis. Reliable biomarkers for MPM diagnosis, monitoring, and prognosis are needed. The aim of this study was to identify genes associated with wound healing processes whose expression could serve as a prognostic factor in MPM patients. Materials and Methods: We used data mining techniques and transcriptomic analysis so as to assess the differential transcriptional expression of wound-healing-associated genes in MPM. Moreover, we investigated the potential prognostic value as well as the functional enrichments of gene ontologies relative to microRNAs (miRNAs) of the significantly differentially expressed wound-healing-related genes in MPM. Results: Out of the 82 wound-healing-associated genes analyzed, 30 were found significantly deregulated in MPM. Kaplan–Meier analysis revealed that low ITGAV gene expression could serve as a prognostic factor favoring survival of MPM patients. Finally, gene ontology annotation enrichment analysis pointed to the members of the hsa-miR-143, hsa-miR-223, and the hsa-miR-29 miRNA family members as important regulators of the deregulated wound healing genes. Conclusions: 30 wound-healing-related genes were significantly deregulated in MPM, which are potential targets of hsa-miR-143, hsa-miR-223, and the hsa-miR-29 miRNA family members. Out of those genes, ITGAV gene expression was a prognostic factor of overall survival in MPM. Our results highlight the role of impaired tissue repair in MPM development and should be further validated experimentally.
Collapse
Affiliation(s)
- Erasmia Rouka
- Department of Transfusion Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
- Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
| | - Eleftherios Beltsios
- Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
| | - Dimos Goundaroulis
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland.
| | | | - Evgeniy I Solenov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia.
- Novosibirsk State University, Novosibirsk 630090, Russia.
| | - Chrissi Hatzoglou
- Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
| | - Konstantinos I Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece.
| |
Collapse
|