1
|
Moran HR, Nyarko OO, O’Rourke R, Ching RCK, Riemslagh FW, Peña B, Burger A, Sucharov CC, Mosimann C. The pericardium forms as a distinct structure during heart formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613484. [PMID: 39345600 PMCID: PMC11429720 DOI: 10.1101/2024.09.18.613484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The heart integrates diverse cell lineages into a functional unit, including the pericardium, a mesothelial sac that supports heart movement, homeostasis, and immune responses. However, despite its critical roles, the developmental origins of the pericardium remain uncertain due to disparate models. Here, using live imaging, lineage tracking, and single-cell transcriptomics in zebrafish, we find the pericardium forms within the lateral plate mesoderm from dedicated anterior mesothelial progenitors and distinct from the classic heart field. Imaging of transgenic reporters in zebrafish documents lateral plate mesoderm cells that emerge lateral of the classic heart field and among a continuous mesothelial progenitor field. Single-cell transcriptomics and trajectories of hand2-expressing lateral plate mesoderm reveal distinct populations of mesothelial and cardiac precursors, including pericardial precursors that are distinct from the cardiomyocyte lineage. The mesothelial gene expression signature is conserved in mammals and carries over to post-natal development. Light sheet-based live-imaging and machine learning-supported cell tracking documents that during heart tube formation, pericardial precursors that reside at the anterior edge of the heart field migrate anteriorly and medially before fusing, enclosing the embryonic heart to form a single pericardial cavity. Pericardium formation proceeds even upon genetic disruption of heart tube formation, uncoupling the two structures. Canonical Wnt/β-catenin signaling modulates pericardial cell number, resulting in a stretched pericardial epithelium with reduced cell number upon canonical Wnt inhibition. We connect the pathological expression of secreted Wnt antagonists of the SFRP family found in pediatric dilated cardiomyopathy to increased pericardial stiffness: sFRP1 in the presence of increased catecholamines causes cardiomyocyte stiffness in neonatal rats as measured by atomic force microscopy. Altogether, our data integrate pericardium formation as an independent process into heart morphogenesis and connect disrupted pericardial tissue properties such as pericardial stiffness to pediatric cardiomyopathies.
Collapse
Affiliation(s)
- Hannah R. Moran
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Obed O. Nyarko
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ryenne-Christine K. Ching
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Frederike W. Riemslagh
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Institute, Division of Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Bioengineering Department, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Fry LM, DenHerder J, Clyde GL, Williams LA, Schneider DA. Biphasic pleural mesothelioma in a goat. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2023; 64:828-832. [PMID: 37663018 PMCID: PMC10426257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
An 8-year-old Saanen goat doe was seen for inappetence, tachycardia, and intermittent bluish-grey discoloration of the oral mucous membranes. On physical examination, the goat was mildly tachypneic and tachycardic, with reduced sounds auscultated on the left side of the thorax. Euthanasia was elected. Necropsy revealed an infiltrative, multinodular mass within the left thoracic cavity and innumerable small, tan nodules disseminated across the pleura of the lungs, thoracic walls, and diaphragm. Upon histologic examination, the mass was composed of highly pleomorphic, fusiform to polygonal cells. Neoplastic cells exhibited positive immunoreactivity for both cytokeratin and vimentin, consistent with a diagnosis of biphasic pleural mesothelioma. Key clinical message: Mesothelioma has rarely been described in the goat but should be considered as a differential diagnosis for thoracic masses in small ruminants, along with thymoma; metastatic neoplasia; carcinomatosis; and granulomatous lesions caused by parasites, bacteria, and fungi.
Collapse
Affiliation(s)
- Lindsay M Fry
- Animal Disease Research Unit, U.S. Department of Agriculture - Agricultural Research Service, Pullman, Washington 99164, USA (Fry, Schneider); Veterinary Microbiology and Pathology Department (Fry, Williams, Schneider) and Office of the Campus Veterinarian (DenHerder, Clyde), Washington State University, Pullman, Washington 99164, USA
| | - Johnathan DenHerder
- Animal Disease Research Unit, U.S. Department of Agriculture - Agricultural Research Service, Pullman, Washington 99164, USA (Fry, Schneider); Veterinary Microbiology and Pathology Department (Fry, Williams, Schneider) and Office of the Campus Veterinarian (DenHerder, Clyde), Washington State University, Pullman, Washington 99164, USA
| | - Gay Lynn Clyde
- Animal Disease Research Unit, U.S. Department of Agriculture - Agricultural Research Service, Pullman, Washington 99164, USA (Fry, Schneider); Veterinary Microbiology and Pathology Department (Fry, Williams, Schneider) and Office of the Campus Veterinarian (DenHerder, Clyde), Washington State University, Pullman, Washington 99164, USA
| | - Laura A Williams
- Animal Disease Research Unit, U.S. Department of Agriculture - Agricultural Research Service, Pullman, Washington 99164, USA (Fry, Schneider); Veterinary Microbiology and Pathology Department (Fry, Williams, Schneider) and Office of the Campus Veterinarian (DenHerder, Clyde), Washington State University, Pullman, Washington 99164, USA
| | - David A Schneider
- Animal Disease Research Unit, U.S. Department of Agriculture - Agricultural Research Service, Pullman, Washington 99164, USA (Fry, Schneider); Veterinary Microbiology and Pathology Department (Fry, Williams, Schneider) and Office of the Campus Veterinarian (DenHerder, Clyde), Washington State University, Pullman, Washington 99164, USA
| |
Collapse
|
3
|
Fasolato S, Del Bianco P, Malacrida S, Mattiolo A, Gringeri E, Angeli P, Pontisso P, Calabrò ML. Studies on the Role of Compartmentalized Profiles of Cytokines in the Risk of Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:13432. [PMID: 37686245 PMCID: PMC10563083 DOI: 10.3390/ijms241713432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common form of liver cancer, is frequently diagnosed late due to the absence of symptoms during early disease, thus heavily affecting the overall survival of these patients. Soluble immunological factors persistently produced during cirrhosis have been recognized as promoters of chronic inflammation and neoplastic transformation. The aim of this pilot study was to evaluate the predictive value of the cytokine profiles for HCC development. A Luminex xMAP approach was used for the quantification of 45 proteins in plasma and ascitic fluids of 44 cirrhotic patients without or with HCC of different etiologies. The association with patient survival was also evaluated. Univariate analyses revealed that very low levels of interleukin 5 (IL-5) (<15.86 pg/mL) in ascites and IL-15 (<12.40 pg/mL) in plasma were able to predict HCC onset with an accuracy of 81.8% and a sensitivity of 95.2%. Univariate analyses also showed that HCC, hepatitis B virus/hepatitis C virus infections, low levels of IL-5 and granulocyte-macrophage colony-stimulating factor in ascitic fluids, and high levels of eotaxin-1, hepatocyte growth factor and stromal-cell-derived factor 1α in plasma samples were factors potentially associated with a poor prognosis and decreased survival. Our results suggest a potential protective role of some immune modulators that may act in the peritoneal cavity to counteract disease progression leading to HCC development.
Collapse
Affiliation(s)
- Silvano Fasolato
- Department of Medicine, Padua University Hospital, I-35128 Padua, Italy; (P.A.); (P.P.)
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy;
| | - Sandro Malacrida
- Institute of Mountain Emergency Medicine, Eurac Research, I-39100 Bozen, Italy;
| | - Adriana Mattiolo
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (A.M.); (M.L.C.)
| | - Enrico Gringeri
- Hepatobiliary Surgery and Liver Transplantation, Padua University Hospital, I-35128 Padua, Italy;
| | - Paolo Angeli
- Department of Medicine, Padua University Hospital, I-35128 Padua, Italy; (P.A.); (P.P.)
| | - Patrizia Pontisso
- Department of Medicine, Padua University Hospital, I-35128 Padua, Italy; (P.A.); (P.P.)
| | - Maria Luisa Calabrò
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, I-35128 Padua, Italy; (A.M.); (M.L.C.)
| |
Collapse
|
4
|
Ries A, Slany A, Pirker C, Mader JC, Mejri D, Mohr T, Schelch K, Flehberger D, Maach N, Hashim M, Hoda MA, Dome B, Krupitza G, Berger W, Gerner C, Holzmann K, Grusch M. Primary and hTERT-Transduced Mesothelioma-Associated Fibroblasts but Not Primary or hTERT-Transduced Mesothelial Cells Stimulate Growth of Human Mesothelioma Cells. Cells 2023; 12:2006. [PMID: 37566084 PMCID: PMC10417280 DOI: 10.3390/cells12152006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023] Open
Abstract
Pleural mesothelioma (PM) is an aggressive malignancy that develops in a unique tumor microenvironment (TME). However, cell models for studying the TME in PM are still limited. Here, we have generated and characterized novel human telomerase reverse transcriptase (hTERT)-transduced mesothelial cell and mesothelioma-associated fibroblast (Meso-CAF) models and investigated their impact on PM cell growth. Pleural mesothelial cells and Meso-CAFs were isolated from tissue of pneumothorax and PM patients, respectively. Stable expression of hTERT was induced by retroviral transduction. Primary and hTERT-transduced cells were compared with respect to doubling times, hTERT expression and activity levels, telomere lengths, proteomes, and the impact of conditioned media (CM) on PM cell growth. All transduced derivatives exhibited elevated hTERT expression and activity, and increased mean telomere lengths. Cell morphology remained unchanged, and the proteomes were similar to the corresponding primary cells. Of note, the CM of primary and hTERT-transduced Meso-CAFs stimulated PM cell growth to the same extent, while CM derived from mesothelial cells had no stimulating effect, irrespective of hTERT expression. In conclusion, all new hTERT-transduced cell models closely resemble their primary counterparts and, hence, represent valuable tools to investigate cellular interactions within the TME of PM.
Collapse
Affiliation(s)
- Alexander Ries
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Astrid Slany
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Johanna C. Mader
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Doris Mejri
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Waehringer Guertel 38, 1090 Vienna, Austria
- ScienceConsult—DI Thomas Mohr KG, Enzianweg 10a, 2353 Guntramsdorf, Austria
| | - Karin Schelch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
| | - Daniela Flehberger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Nadine Maach
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Muhammad Hashim
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
| | - Balazs Dome
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
- National Korányi Institute of Pulmonology, Korányi Frigyes u. 1, 1122 Budapest, Hungary
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary
- Department of Translational Medicine, Lund University, Sölvegatan 19, 22184 Lund, Sweden
| | - Georg Krupitza
- Department of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Klaus Holzmann
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Michael Grusch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| |
Collapse
|
5
|
Metelmann IB, Kraemer S, Steinert M, Langer S, Stock P, Kurow O. Novel 3D organotypic co-culture model of pleura. PLoS One 2022; 17:e0276978. [PMID: 36454800 PMCID: PMC9714887 DOI: 10.1371/journal.pone.0276978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Pleural mesothelial cells are the predominant cell type in the pleural cavity, but their role in the pathogenesis of pleural diseases needs to be further elucidated. 3D organotypic models are an encouraging approach for an in vivo understanding of molecular disease development. The aim of the present study was to develop a 3D organotypic model of the pleural mesothelium. Specimens of human pleura parietalis were obtained from patients undergoing surgery at the University Hospital Leipzig, Germany. 3D co-culture model of pleura was established from human pleural mesothelial cells and fibroblasts. The model was compared to human pleura tissue by phase-contrast and light microscopy, immunochemistry and -fluorescence as well as solute permeation test. Histological assessment of the 3D co-culture model displayed the presence of both cell types mimicking the morphology of the human pleura. Vimentin and Cytokeratin, PHD1 showed a similar expression pattern in pleural biopsies and 3D model. Expression of Ki-67 indicates the presence of proliferating cells. Tight junctional marker ZO-1 was found localized at contact zones between mesothelial cells. Each of these markers were expressed in both the 3D co-culture model and human biopsies. Permeability of 3D organotypic co-culture model of pleura was found to be higher for 70 kDa-Dextran and no significant difference was seen in the permeability for small dextran (4 kDa). In summary, the presented 3D organoid of pleura functions as a robust assay for pleural research serving as a precise reproduction of the in vivo morphology and microenvironment.
Collapse
Affiliation(s)
- Isabella B. Metelmann
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Sebastian Kraemer
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Matthias Steinert
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Stefan Langer
- Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Peggy Stock
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Olga Kurow
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
6
|
Visser D, Bakhshi H, Rogg K, Fuhrmann E, Wieland F, Schenke-Layland K, Meyer W, Hartmann H. Green Chemistry for Biomimetic Materials: Synthesis and Electrospinning of High-Molecular-Weight Polycarbonate-Based Nonisocyanate Polyurethanes. ACS OMEGA 2022; 7:39772-39781. [PMID: 36385898 PMCID: PMC9648058 DOI: 10.1021/acsomega.2c03731] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
Conventional synthesis routes for thermoplastic polyurethanes (TPUs) still require the use of isocyanates and tin-based catalysts, which pose considerable safety and environmental hazards. To reduce both the ecological footprint and human health dangers for nonwoven TPU scaffolds, it is key to establish a green synthesis route, which eliminates the use of these toxic compounds and results in biocompatible TPUs with facile processability. In this study, we developed high-molecular-weight nonisocyanate polyurethanes (NIPUs) through transurethanization of 1,6-hexanedicarbamate with polycarbonate diols (PCDLs). Various molecular weights of PCDL were employed to maximize the molecular weight of NIPUs and consequently facilitate their electrospinnability. The synthesized NIPUs were characterized by nuclear magnetic resonance, Fourier-transform infrared spectroscopy, gel permeation chromatography, and differential scanning calorimetry. The highest achieved molecular weight (M w) was 58,600 g/mol. The NIPUs were consecutively electrospun into fibrous scaffolds with fiber diameters in the submicron range, as shown by scanning electron microscopy (SEM). To assess the suitability of electrospun NIPU mats as a possible biomimetic load-bearing pericardial substitute in cardiac tissue engineering, their cytotoxicity was investigated in vitro using primary human fibroblasts and a human epithelial cell line. The bare NIPU mats did not need further biofunctionalization to enhance cell adhesion, as it was not outperformed by collagen-functionalized NIPU mats and hence showed that the NIPU mats possess a great potential for use in biomimetic scaffolds.
Collapse
Affiliation(s)
- Dmitri Visser
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Hadi Bakhshi
- Department
of Life Science and Bioprocesses, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Katharina Rogg
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Ellena Fuhrmann
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| | - Franziska Wieland
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Katja Schenke-Layland
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
- Institute
of Biomedical Engineering, Dept. for Medical Technologies and Regenerative
Medicine, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Cluster
of Excellence iFIT (EXC 2180) “Image-Guided and Functionally
Instructed Tumor Therapies”, Eberhard
Karls University Tübingen, 72076 Tübingen, Germany
| | - Wolfdietrich Meyer
- Department
of Life Science and Bioprocesses, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
- Department
of Functional Polymer Systems, Fraunhofer
Institute for Applied Polymer Research IAP, Geiselbergstraße 69, 14476 Potsdam, Germany
| | - Hanna Hartmann
- NMI
Natural and Medical Science Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Germany
| |
Collapse
|
7
|
Yan Z, Zhang Y, Wang C, Li Y, Su Q, Cao J, Cao X. Withaferin a Attenuates Retinal Ischemia-Reperfusion Injury via Akt-Dependent Inhibition of Oxidative Stress. Cells 2022; 11:cells11193113. [PMID: 36231074 PMCID: PMC9563317 DOI: 10.3390/cells11193113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Retinal ischemia-reperfusion (I/R) injury often results in intractable visual impairments. The survival of retinal capillary endothelial cells is crucial for the treatment of retinal I/R injury. How to protect retinal endothelia from damage is a challenging work. Withaferin A, a small molecule derived from plants, has antibacterial and anti-inflammatory effects and has been used for about millennia in traditional medicine. The present study aimed to investigate the potential protective effect of withaferin A on retinal I/R injury. Methods: The drug-likeness of withaferin A was evaluated by the SwissADME web tool. The potential protective effect of withaferin A on the I/R-induced injury of human retinal microvascular endothelial cells (HRMECs) was investigated using multiple approaches. RNA sequencing was performed and associated mechanistic signaling pathways were analyzed based on the Kyoto Encyclopedia of Genes and Genomes data. The analytical results of RNA sequencing data were further validated by in vitro and in vivo experiments. Results: Withaferin A reduced the I/R injury-induced apoptotic death of HRMECs in vitro with a good drug-like property. RNA sequencing and experimental validation results indicated that withaferin A increased the production of the crucial antioxidant molecules heme oxygenase 1 (HO-1) and peroxiredoxin 1 (Prdx-1) during I/R. In addition, withaferin A activated the Akt signaling pathway and increased the expression of HO-1 and Prdx-1, thereby exerting an antioxidant effect, attenuated the retinal I/R injury, and decreased the apoptosis of HRMECs. The blockade of Akt completely abolished the effects of withaferin A. Conclusions: The study identified for the first time that withaferin A can protect against the I/R-induced apoptosis of human microvascular retinal endothelial cells via increasing the production of the antioxidants Prdx-1 and HO-1. Results suggest that withaferin A is a promising drug candidate for the treatment of retinal I/R injury.
Collapse
Affiliation(s)
- Zheyi Yan
- Department of Ophthalmology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yuanlin Zhang
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan 030619, China
| | - Chunfang Wang
- Department of Ophthalmology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yanjie Li
- Department of Ophthalmology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Qiang Su
- Department of Ophthalmology, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoming Cao
- Department of Orthopedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan 030000, China
- Correspondence:
| |
Collapse
|
8
|
Polycaprolactone/Chitosan Composite Nanofiber Membrane as a Preferred Scaffold for the Culture of Mesothelial Cells and the Repair of Damaged Mesothelium. Int J Mol Sci 2022; 23:ijms23179517. [PMID: 36076916 PMCID: PMC9455682 DOI: 10.3390/ijms23179517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/26/2022] Open
Abstract
Mesothelial cells are specific epithelial cells lining the serosal cavity and internal organs. Nonetheless, few studies have explored the possibility to culture mesothelial cells in a nanostructure scaffold for tissue engineering applications. Therefore, this study aims to fabricate nanofibers from a polycaprolactone (PCL) and PCL/chitosan (CS) blend by electrospinning, and to elucidate the effect of CS on the cellular response of mesothelial cells. The results demonstrate that a PCL and PCL/CS nanofiber membrane scaffold could be prepared with a comparable fiber diameter (~300 nm) and porosity for cell culture. Blending CS with PCL influenced the mechanical properties of the scaffold due to interference of PCL crystallinity in the nanofibers. However, CS substantially improves scaffold hydrophilicity and results in a ~6-times-higher cell attachment rate in PCL/CS. The mesothelial cells maintain high viability in both nanofiber membranes, but PCL/CS provides better maintenance of cobblestone-like mesothelial morphology. From gene expression analysis and immunofluorescence staining, the incorporation of CS also results in the upregulated expression of mesothelial marker genes and the enhanced production of key mesothelial maker proteins, endorsing PCL/CS to better maintain the mesothelial phenotype. The PCL/CS scaffold was therefore chosen for the in vivo studies, which involved transplanting a cell/scaffold construct containing allograft mesothelial cells for mesothelium reconstruction in rats. In the absence of mesothelial cells, the mesothelium wound covered with PCL/CS showed an inflammatory response. In contrast, a mesothelium layer similar to native mesothelium tissue could be obtained by implanting the cell/scaffold construct, based on hematoxylin and eosin (H&E) and immunohistochemical staining.
Collapse
|
9
|
Human Omental Mesothelial Cells Impart an Immunomodulatory Landscape Impeding B- and T-Cell Activation. Int J Mol Sci 2022; 23:ijms23115924. [PMID: 35682603 PMCID: PMC9180401 DOI: 10.3390/ijms23115924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
Mesothelial cells form the mesothelium, a simple epithelium lining the walls of serous cavities and the surface of visceral organs. Although mesothelial cells are phenotypically well characterized, their immunoregulatory properties remain largely unknown, with only two studies reporting their capacity to inhibit T cells through TGF-β and their consumption of L-arginine by arginase-1. Whether human mesothelial cells can suppress other immune cells and possess additional leukosuppressive mechanisms, remain to be addressed to better delineate their therapeutic potential for cell therapy. Herein, we generated secretomes from omental mesothelial cells (OMC) and assess their capacity to inhibit lymphocytes proliferation, suppress activated T and B cells, as well as to modify macrophage activation markers. The secretome from mesenchymal stromal cells (MSC) served as a control of immuno-suppression. Although OMC and MSC were phenotypically divergent, their cytokine secretion patterns as well as expression of inflammatory and immunomodulary genes were similar. As such, OMC- and MSC-derived secretomes (OMC-S and MSC-S) both polarized RAW 264.7 macrophages towards a M2-like anti-inflammatory phenotype and suppressed mouse and human lymphocytes proliferation. OMC-S displayed a strong ability to suppress mouse- and human-activated CD19+/CD25+ B cells as compared to MSC-S. The lymphosuppressive activity of the OMC-S could be significantly counteracted either by SB-431542, an inhibitor of TGFβ and activin signaling pathways, or with a monoclonal antibody against the TGFβ1, β2, and β3 isoforms. A strong blockade of the OMC-S-mediated lymphosuppressive activity was achieved using L-NMMA, a specific inhibitor of nitric oxide synthase (NOS). Taken together, our results suggest that OMC are potent immunomodulators.
Collapse
|
10
|
Haghshenas M, Tavana S, Zand E, Montazeri L, Fathi R. Mouse ovarian follicle growth in an amniotic membrane-based hydrogel. J Biomater Appl 2022; 37:563-574. [PMID: 35451867 DOI: 10.1177/08853282221094193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Three-dimensional cultures of follicles on ECM-based scaffolds can be an approach for women who become infertile after cancer treatments. Human amniotic membrane (HAM) is extensively employed in tissue engineering because of its unique properties. We cultured mouse pre-antral follicles in a hydrogel derived from decellularized amniotic membrane (DAM) combined with alginate (ALG) to improve ovarian follicle culture. HAM was decellularized. Quantitative (nuclear contents, collagen, glycosaminoglycan [GAG]) and qualitative (DAPI, H&E, Masson's trichrome, Alcian blue, scanning electron microscopy assessments were performed. Then, we created an amniotic membrane-based hydrogel (AMBH) and conducted AMBH characterization assays (rheology, MTS, degradation rate). Isolated mouse pre-antral follicles were cultured in 15 mg/mL AMBH (AMBH15), 30 mg/mL AMBH (AMBH30), or 45 mg/mL AMBH (AMBH45). ALG hydrogel was the control group. Follicular diameters, estradiol hormone secretion rate, follicular morphology, and the follicle antral and degeneration rate were examined. Quantitative and qualitative assays indicated successful decellularization. AMBH characterization assays showed that the ALG hydrogel had more appropriate gelation and slower degradation than AMBH. There was a statistically higher antral follicle formation rate in the AMBH45 group (p < .05) compared to the AMBH30 and AMBH15 groups and less (p < .05) degenerated follicles. There was no significant difference with the ALG group. Diameter and estradiol hormone secretion in the AMBH45 group were not significantly higher than the ALG group. Although decellularization was confirmed and the viscoelastic parameters of AMBH support follicle culture, there was no significant effect on ovarian follicle maturation compared to the ALG control group.
Collapse
Affiliation(s)
- Mohammad Haghshenas
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,48499Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Somayeh Tavana
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Elnaz Zand
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Rouhollah Fathi
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
11
|
Huang W, Li X, Huang C, Tang Y, Zhou Q, Chen W. LncRNAs and Rheumatoid Arthritis: From Identifying Mechanisms to Clinical Investigation. Front Immunol 2022; 12:807738. [PMID: 35087527 PMCID: PMC8786719 DOI: 10.3389/fimmu.2021.807738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic chronic autoinflammatory disease, and the synovial hyperplasia, pannus formation, articular cartilage damage and bone matrix destruction caused by immune system abnormalities are the main features of RA. The use of Disease Modifying Anti-Rheumatic Drugs (DMARDs) has achieved great advances in the therapy of RA. Yet there are still patients facing the problem of poor response to drug therapy or drug intolerance. Current therapy methods can only moderate RA progress, but cannot stop or reverse the damage it has caused. Recent studies have reported that there are a variety of long non-coding RNAs (LncRNAs) that have been implicated in mediating many aspects of RA. Understanding the mechanism of LncRNAs in RA is therefore critical for the development of new therapy strategies and prevention strategies. In this review, we systematically elucidate the biological roles and mechanisms of action of LncRNAs and their mechanisms of action in RA. Additionally, we also highlight the potential value of LncRNAs in the clinical diagnosis and therapy of RA.
Collapse
Affiliation(s)
- Wentao Huang
- Ministry of Education (MOE) Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xue Li
- Ministry of Education (MOE) Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Chen Huang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central, Hospital, Guangzhou, China
| | - Yukuan Tang
- Department of Minimally Invasive Interventional Radiology, Guangzhou Panyu Central, Hospital, Guangzhou, China
| | - Quan Zhou
- Department of Radiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wenli Chen
- Ministry of Education (MOE) Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China.,Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
12
|
Capeling MM, Huang S, Childs CJ, Wu JH, Tsai YH, Wu A, Garg N, Holloway EM, Sundaram N, Bouffi C, Helmrath M, Spence JR. Suspension culture promotes serosal mesothelial development in human intestinal organoids. Cell Rep 2022; 38:110379. [PMID: 35172130 PMCID: PMC9002973 DOI: 10.1016/j.celrep.2022.110379] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 09/08/2021] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Pluripotent-stem-cell-derived human intestinal organoids (HIOs) model some aspects of intestinal development and disease, but current culture methods do not fully recapitulate the diverse cell types and complex organization of the human intestine and are reliant on 3D extracellular matrix or hydrogel systems, which limit experimental control and translational potential for regenerative medicine. We describe suspension culture as a simple, low-maintenance method for culturing HIOs and for promoting in vitro differentiation of an organized serosal mesothelial layer that is similar to primary human intestinal serosal mesothelium based on single-cell RNA sequencing and histological analysis. Functionally, HIO serosal mesothelium has the capacity to differentiate into smooth-muscle-like cells and exhibits fibrinolytic activity. An inhibitor screen identifies Hedgehog and WNT signaling as regulators of human serosal mesothelial differentiation. Collectively, suspension HIOs represent a three-dimensional model to study the human serosal mesothelium.
Collapse
Affiliation(s)
- Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Charlie J Childs
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joshua H Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Neil Garg
- School of Kinesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nambirajan Sundaram
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Carine Bouffi
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Michael Helmrath
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Jason R Spence
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Patel SK, Valicherla GR, Micklo AC, Rohan LC. Drug delivery strategies for management of women's health issues in the upper genital tract. Adv Drug Deliv Rev 2021; 177:113955. [PMID: 34481034 DOI: 10.1016/j.addr.2021.113955] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/23/2021] [Accepted: 08/28/2021] [Indexed: 02/08/2023]
Abstract
The female upper genital tract (UGT) hosts important reproductive organs including the cervix, uterus, fallopian tubes, and ovaries. Several pathologies affect these organ systems such as infections, reproductive issues, structural abnormalities, cancer, and inflammatory diseases that could have significant impact on women's overall health. Effective disease management is constrained by the multifaceted nature of the UGT, complex anatomy and a dynamic physiological environment. Development of drug delivery strategies that can overcome mucosal and safety barriers are needed for effective disease management. This review introduces the anatomy, physiology, and mucosal properties of the UGT and describes drug delivery barriers, advances in drug delivery technologies, and opportunities available for new technologies that target the UGT.
Collapse
|
14
|
Huang H, Kuang X, Zhu X, Cheng H, Zou Y, Du H, Tang H, Zhou L, Zeng J, Liu H, Yan J, Long C, Shen H. Maintaining blood retinal barrier homeostasis to attenuate retinal ischemia-reperfusion injury by targeting the KEAP1/NRF2/ARE pathway with lycopene. Cell Signal 2021; 88:110153. [PMID: 34571190 DOI: 10.1016/j.cellsig.2021.110153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/15/2022]
Abstract
Retinal ischemia-reperfusion (I/R) often results in intractable visual impairments, where blood retinal barrier (BRB) homeostasis mediated by retinal pigment epithelium (RPE) and retinal microvascular endothelium (RME) is crucial. However, strategies targeting the BRB are limited. Thus, we investigated the inconclusive effect of lycopene (LYC) in retinal protection under I/R. LYC elevated cellular viability and reversed oxidative stress in aRPE-19 cells/hRME cells under I/R conditions based on oxygen-glucose deprivation (OGD) in vitro. Molecular analysis showed that LYC promoted NRF2 expression and enhanced the downstream factors of the KEAP1/NRF2/ARE pathway: LYC increased the activities of antioxidants, including SOD and CAT, whereas it enhanced the mRNA expression of HO-1 (ho-1) and NQO-1 (nqo-1). The activation resulted in restrained ROS and MDA. On the other hand, LYC ameliorated the damage to retinal function and morphology in a mouse I/R model, which was established by unilateral ligation of the left pterygopalatine artery/external carotid artery and reperfusion. LYC promoted the expression of NRF2 in both the neural retina and the RPE choroid in vivo. This evidence revealed the potential of LYC in retinal protection under I/R, uncovering the pharmacological effect of the KEAP1/NRF2/ARE pathway in BRB targeting. The study generates new insights into scientific practices in retinal research.
Collapse
Affiliation(s)
- Hao Huang
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; Department of Ophthalmology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xielan Kuang
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaobo Zhu
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Hao Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuxiu Zou
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Han Du
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Han Tang
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Linbin Zhou
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jingshu Zeng
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Huijun Liu
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jianhua Yan
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chongde Long
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Huangxuan Shen
- State Key laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China; Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
15
|
Sarabadani M, Tavana S, Mirzaeian L, Fathi R. Co-culture with peritoneum mesothelial stem cells supports the in vitro growth of mouse ovarian follicles. J Biomed Mater Res A 2021; 109:2685-2694. [PMID: 34228401 DOI: 10.1002/jbm.a.37260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
The important roles played by the ovarian microenvironment and cell interactions in folliculogenesis suggest promising approaches for in vivo growth of ovarian follicles using appropriate scaffolds containing suitable cell sources. In this study, we have investigated the growth of early preantral follicles in the presence of decellularized mesenteric peritoneal membrane (MPM), peritoneum mesothelial stem cells (PMSCs), and conditioned medium (CM) of PMSCs. MPM of mouse was first decellularized; PMSCs were isolated from MPM and cultured and their conditioned medium (CM) was collected. Mouse follicles were separated into four groups: (1) culture in base medium (control), (2) culture in decellularized MPM (DMPM), (3) co-culture with PMSCs (Co-PMSCs), and (4) culture in CM of PMSCs (CM-PMSCs). Qualitative and quantitative assessments were performed to evaluate intact mesenteric peritoneal membrane (IMPM) as well as decellularized ones. After culturing the ovarian follicles, follicular and oocyte diameter, viability, eccentric oocyte percentage, and estradiol hormone amounts were evaluated. Quantitative and qualitative evaluations confirmed removal of cells and retention of the essential fibers in MPM after the decellularization process. Follicular parameters showed that Co-PMSCs better support in vitro growth and development of ovarian follicles than the other groups. The eccentric rate and estradiol production were statistically higher for the Co-PMSCs group than for the CM-PMSCs and control groups. Although the culture of early preantral follicles on DMPM and CM-PMSCs could improve in vitro follicular growth, co-culture of follicles with PMSCs showed even greater improvements in terms of follicular growth and diameter.
Collapse
Affiliation(s)
- Mahdieh Sarabadani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Leila Mirzaeian
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
16
|
Crohn's Disease Increases the Mesothelial Properties of Adipocyte Progenitors in the Creeping Fat. Int J Mol Sci 2021; 22:ijms22084292. [PMID: 33924264 PMCID: PMC8074767 DOI: 10.3390/ijms22084292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Our understanding of the interplay between human adipose tissue and the immune system is limited. The mesothelium, an immunologically active structure, emerged as a source of visceral adipose tissue. After investigating the mesothelial properties of human visceral and subcutaneous adipose tissue and their progenitors, we explored whether the dysfunctional obese and Crohn's disease environments influence the mesothelial/mesenchymal properties of their adipocyte precursors, as well as their ability to mount an immune response. Using a tandem transcriptomic/proteomic approach, we evaluated the mesothelial and mesenchymal expression profiles in adipose tissue, both in subjects covering a wide range of body-mass indexes and in Crohn's disease patients. We also isolated adipose tissue precursors (adipose-derived stem cells, ASCs) to assess their mesothelial/mesenchymal properties, as well as their antigen-presenting features. Human visceral tissue presented a mesothelial phenotype not detected in the subcutaneous fat. Only ASCs from mesenteric adipose tissue, named creeping fat, had a significantly higher expression of the hallmark mesothelial genes mesothelin (MSLN) and Wilms' tumor suppressor gene 1 (WT1), supporting a mesothelial nature of these cells. Both lean and Crohn's disease visceral ASCs expressed equivalent surface percentages of the antigen-presenting molecules human leucocyte antigen-DR isotype (HLA-DR) and CD86. However, lean-derived ASCs were predominantly HLA-DR dim, whereas in Crohn's disease, the HLA-DR bright subpopulation was increased 3.2-fold. Importantly, the mesothelial-enriched Crohn's disease precursors activated CD4+ T-lymphocytes. Our study evidences a mesothelial signature in the creeping fat of Crohn's disease patients and its progenitor cells, the latter being able to present antigens and orchestrate an immune response.
Collapse
|
17
|
Chen B, Chen X, Wang W, Shen J, Song Z, Ji H, Zhang F, Wu J, Na J, Li S. Tissue-engineered autologous peritoneal grafts for bladder reconstruction in a porcine model. J Tissue Eng 2021; 12:2041731420986796. [PMID: 33613958 PMCID: PMC7874343 DOI: 10.1177/2041731420986796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/18/2020] [Indexed: 01/08/2023] Open
Abstract
Ileal neobladder construction is a common treatment for patients with bladder cancer after radical cystectomy. However, metabolic disorders caused by transposed bowel segments occur frequently. Bladder tissue engineering is a promising alternative approach. Although numerous studies have reported bladder reconstruction using acellular and cellular scaffolds, there are also disadvantages associated with these methods, such as immunogenicity of synthetic grafts and incompatible mechanical properties of the biomaterials. Here, we engineered an autologous peritoneal graft consisting of a peritoneal sheet and the seromuscular layer from the ileum. Three months after the surgery, compared with the neobladder made from the ileum, the reconstructed neobladder using our new method showed normal function and better gross morphological characteristics. Moreover, histopathological and transcriptomic analysis revealed urothelium-like cells expressing urothelial biomarkers appeared in the neobladder, while no such changes were observed in the control group. Overall, our study provides a new strategy for bladder tissue engineering and informs a variety of future research prospects.
Collapse
Affiliation(s)
- Biao Chen
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Xia Chen
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Wenjia Wang
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jun Shen
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhiqiang Song
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Haoyu Ji
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Fangyuan Zhang
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jianchen Wu
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jie Na
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Shengwen Li
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| |
Collapse
|
18
|
Abstract
The skin is the largest organ in the body, fulfilling a variety of functions and acting as a barrier for internal organs against external insults. As for extensive or irreversible damage, skin autografts are often considered the gold standard, however inherent limitations highlight the need for alternative strategies. Engineering of human-compatible tissues is an interdisciplinary and active field of research, leading to the production of scaffolds and skin substitutes to guide repair and regeneration. However, faithful reproduction of extracellular matrix (ECM) architecture and bioactive content capable of cell-instructive and cell-responsive properties remains challenging. ECM is a heterogeneous, connective network composed of collagens, glycoproteins, proteoglycans, and small molecules. It is highly coordinated to provide the physical scaffolding, mechanical stability, and biochemical cues necessary for tissue morphogenesis and homeostasis. Decellularization processes have made it possible to isolate the ECM in its native and three-dimensional form from a cell-populated tissue for use in skin regeneration. In this review, we present recent knowledge about these decellularized biomaterials with the potential to be used as dermal or skin substitutes in clinical applications. We detail tissue sources and clinical indications with success rates and report the most effective decellularization methods compatible with clinical use.
Collapse
|
19
|
Ramírez-Hernández C, García-Márquez LJ, Decanini-Arcaute H, Martínez-Burnes J, Ramírez-Romero R. Fat, Cartilage, and Bone Metaplasia in Lungs of Cattle With Caudal Pleural Lesions and Subjacent Interstitial Fibrosis. Vet Pathol 2019; 56:599-603. [PMID: 30917746 DOI: 10.1177/0300985819837719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The changes associated with condemned lungs in cattle with chronic pleural lesions of the caudal lobes were characterized by histology and immunohistochemistry (IHC). Fibroproliferative pleural lesions were microscopically confirmed. Occasionally, the pleural lesions also included adipose, chondroid, and osseous metaplasia that were covered by mesothelial cells, mostly in the absence of inflammation. Other lungs also showed fibrosis in the subpleural interstitium and interlobular septa. In both condemned and noncondemned lungs, immunoreactivity to Wilms tumor 1 (WT1) was normally observed on surface mesothelial cells but not on the submesothelial fibroblasts and myofibroblasts. Conversely, the myofibroblasts beneath the pleura, but not the mesothelial cells, showed immunoreactivity to alpha smooth muscle actin and calponin. However, in the lungs with myofibroblastic foci in the pleura, the proliferated cells maintained WT1 immunoreactivity similar to those of some metaplastic cells. These findings may reflect the plasticity of mesothelial cells in vivo.
Collapse
Affiliation(s)
- Cecilia Ramírez-Hernández
- 1 Universidad Autónoma de Nuevo León, Posgrado Conjunto Agronomía-Veterinaria, Gral. Escobedo, Nuevo Leon, México
| | - Luis Jorge García-Márquez
- 2 Centro Universitario de Investigación y Desarrollo Agropecuario (CUIDA), Facultad de Medicina Veterinaria y Zootecnia, Universidad de Colima, México
| | - Horacio Decanini-Arcaute
- 3 Departamento de Patología, Hospital Christus-Muguerza Alta Especialidad, Monterrey, Nuevo Leon, México
| | - Julio Martínez-Burnes
- 4 Facultad de Medicina Veterinaria y Zootecnia "Dr. Norberto Treviño Zapata", Universidad Autónoma de Tamaulipas, México
| | - Rafael Ramírez-Romero
- 1 Universidad Autónoma de Nuevo León, Posgrado Conjunto Agronomía-Veterinaria, Gral. Escobedo, Nuevo Leon, México
| |
Collapse
|
20
|
Kress S, Baur J, Otto C, Burkard N, Braspenning J, Walles H, Nickel J, Metzger M. Evaluation of a Miniaturized Biologically Vascularized Scaffold in vitro and in vivo. Sci Rep 2018; 8:4719. [PMID: 29549334 PMCID: PMC5856827 DOI: 10.1038/s41598-018-22688-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 02/28/2018] [Indexed: 02/07/2023] Open
Abstract
In tissue engineering, the generation and functional maintenance of dense voluminous tissues is mainly restricted due to insufficient nutrient supply. Larger three-dimensional constructs, which exceed the nutrient diffusion limit become necrotic and/or apoptotic in long-term culture if not provided with an appropriate vascularization. Here, we established protocols for the generation of a pre-vascularized biological scaffold with intact arterio-venous capillary loops from rat intestine, which is decellularized under preservation of the feeding and draining vascular tree. Vessel integrity was proven by marker expression, media/blood reflow and endothelial LDL uptake. In vitro maintenance persisted up to 7 weeks in a bioreactor system allowing a stepwise reconstruction of fully vascularized human tissues and successful in vivo implantation for up to 4 weeks, although with time-dependent decrease of cell viability. The vascularization of the construct lead to a 1.5× increase in cellular drug release compared to a conventional static culture in vitro. For the first time, we performed proof-of-concept studies demonstrating that 3D tissues can be maintained within a miniaturized vascularized scaffold in vitro and successfully implanted after re-anastomosis to the intrinsic blood circulation in vivo. We hypothesize that this technology could serve as a powerful platform technology in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sebastian Kress
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany
| | - Johannes Baur
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Christoph Otto
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Natalie Burkard
- University Hospital of Würzburg, Department of General, Visceral, Vascular and Pediatric Surgery, 97080, Würzburg, Germany
| | - Joris Braspenning
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany
| | - Heike Walles
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany.,Fraunhofer Institute of Silicate Research ISC, Translational Center for Regenerative Therapies, 97070, Würzburg, Germany
| | - Joachim Nickel
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany.
| | - Marco Metzger
- University Hospital of Würzburg, Chair of Tissue Engineering and Regenerative Medicine, 97070, Würzburg, Germany. .,Fraunhofer Institute of Silicate Research ISC, Translational Center for Regenerative Therapies, 97070, Würzburg, Germany.
| |
Collapse
|
21
|
Straka F, Schornik D, Masin J, Filova E, Mirejovsky T, Burdikova Z, Svindrych Z, Chlup H, Horny L, Daniel M, Machac J, Skibová J, Pirk J, Bacakova L. A human pericardium biopolymeric scaffold for autologous heart valve tissue engineering: cellular and extracellular matrix structure and biomechanical properties in comparison with a normal aortic heart valve. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:599-634. [PMID: 29338582 DOI: 10.1080/09205063.2018.1429732] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The objective of our study was to compare the cellular and extracellular matrix (ECM) structure and the biomechanical properties of human pericardium (HP) with the normal human aortic heart valve (NAV). HP tissues (from 12 patients) and NAV samples (from 5 patients) were harvested during heart surgery. The main cells in HP were pericardial interstitial cells, which are fibroblast-like cells of mesenchymal origin similar to the valvular interstitial cells in NAV tissue. The ECM of HP had a statistically significantly (p < 0.001) higher collagen I content, a lower collagen III and elastin content, and a similar glycosaminoglycans (GAGs) content, in comparison with the NAV, as measured by ECM integrated density. However, the relative thickness of the main load-bearing structures of the two tissues, the dense part of fibrous HP (49 ± 2%) and the lamina fibrosa of NAV (47 ± 4%), was similar. In both tissues, the secant elastic modulus (Es) was significantly lower in the transversal direction (p < 0.05) than in the longitudinal direction. This proved that both tissues were anisotropic. No statistically significant differences in UTS (ultimate tensile strength) values and in calculated bending stiffness values in the longitudinal or transversal direction were found between HP and NAV. Our study confirms that HP has an advantageous ECM biopolymeric structure and has the biomechanical properties required for a tissue from which an autologous heart valve replacement may be constructed.
Collapse
Affiliation(s)
- Frantisek Straka
- a Cardiology Centre and Cardiovascular Surgery Department , Institute for Clinical and Experimental Medicine , Prague , Czech Republic.,b Department of Biomaterials and Tissue Engineering , Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - David Schornik
- b Department of Biomaterials and Tissue Engineering , Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Jaroslav Masin
- a Cardiology Centre and Cardiovascular Surgery Department , Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | - Elena Filova
- b Department of Biomaterials and Tissue Engineering , Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| | - Tomas Mirejovsky
- c Clinical and Transplant Pathology Department, Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | - Zuzana Burdikova
- d Department of Cell Biology, School of Medicine , University of Virginia , Charlottesville , VA , USA
| | - Zdenek Svindrych
- e Department of Biology, W. M, Keck Center for Cellular Imaging , University of Virginia , Charlottesville , VA , USA
| | - Hynek Chlup
- f Faculty of Mechanical Engineering, Department of Mechanics, Biomechanics and Mechatronics , Czech Technical University in Prague , Prague , Czech Republic
| | - Lukas Horny
- f Faculty of Mechanical Engineering, Department of Mechanics, Biomechanics and Mechatronics , Czech Technical University in Prague , Prague , Czech Republic
| | - Matej Daniel
- f Faculty of Mechanical Engineering, Department of Mechanics, Biomechanics and Mechatronics , Czech Technical University in Prague , Prague , Czech Republic
| | - Jiri Machac
- g Institute of Botany CAS, Academy of Sciences of the Czech Republic , Pruhonice , Czech Republic
| | - Jelena Skibová
- h Department of Medical Statistics , Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | - Jan Pirk
- a Cardiology Centre and Cardiovascular Surgery Department , Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | - Lucie Bacakova
- b Department of Biomaterials and Tissue Engineering , Institute of Physiology, Academy of Sciences of the Czech Republic , Prague , Czech Republic
| |
Collapse
|
22
|
Mutsaers SE, Prêle CMA, Pengelly S, Herrick SE. Mesothelial cells and peritoneal homeostasis. Fertil Steril 2017; 106:1018-1024. [PMID: 27692285 DOI: 10.1016/j.fertnstert.2016.09.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
The mesothelium was traditionally thought to be a simple tissue with the sole function of providing a slippery, nonadhesive, and protective surface to allow easy movement of organs within their body cavities. However, our knowledge of mesothelial cell physiology is rapidly expanding, and the mesothelium is now recognized as a dynamic cellular membrane with many other important functions. When injured, mesothelial cells initiate a cascade of processes leading either to complete regeneration of the mesothelium or the development of pathologies such as adhesions. Normal mesothelial healing is unique in that, unlike with other epithelial-like surfaces, healing appears diffusely across the denuded surface, whereas for epithelium healing occurs solely at the wound edges. This is because of a free-floating population of mesothelial cells which attach to the injured serosa. Taking advantage of this phenomenon, intraperitoneal injections of mesothelial cells have been assessed for their ability to prevent adhesion formation. This review discusses some of the functions of mesothelial cells regarding maintenance of serosal integrity and outlines the mechanisms involved in mesothelial healing. In addition, the pathogenesis of adhesion formation is discussed with particular attention to the potential role of mesothelial cells in both preventing and inducing their development.
Collapse
Affiliation(s)
- Steven Eugene Mutsaers
- Institute for Respiratory Health, Centre for Respiratory Health, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.
| | - Cecilia Marie-Antoinette Prêle
- Institute for Respiratory Health, Centre for Respiratory Health, and Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | - Steven Pengelly
- Institute of Inflammation and Repair, Faculty of Medical and Human Sciences and Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Sarah Elizabeth Herrick
- Institute of Inflammation and Repair, Faculty of Medical and Human Sciences and Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
23
|
Kawanishi K. Mesothelial cell transplantation: history, challenges and future directions. Pleura Peritoneum 2016; 1:135-143. [PMID: 30911617 PMCID: PMC6419540 DOI: 10.1515/pp-2016-0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022] Open
Abstract
Mesothelial cells line the surface of the pleura, pericardium, peritoneum and internal reproductive organs. One of their main functions is to act as a non-adhesive barrier to protect against physical damage, however, over the past decades their physiological and pathological properties have been revealed in association with a variety of conditions and diseases. Mesothelium has been used in surgical operations in clinical settings, such as omental patching for perforated peptic ulcers and in glutaraldehyde-treated autologous pericardium for aortic valve reconstruction. Various methods for mesothelial cell transplantation have also been established and developed, particularly within the area of tissue engineering, including scaffold and non-scaffold cell sheet technologies. However, the use of mesothelial cell transplantation in patients remains challenging, as it requires additional operations under general anesthesia in order to obtain enough intact cells for culture. Moreover, the current methods of mesothelial cell transplantation are expensive and are not yet available in clinical practice. This review firstly summarizes the history of the use of mesothelial cell transplantation in tissue engineering, and then critically discusses the barriers for the clinical application of mesothelial cell transplantation. Finally, the recent developments in xenotransplantation technologies are discussed to evaluate other feasible alternatives to mesothelial cell transplantation.
Collapse
Affiliation(s)
- Kunio Kawanishi
- Department of Cellular and Molecular Medicine, University of California, San Diego,9500 Gilman Drive, La Jolla, CA 92093–0687, USA
- Department of Surgical Pathology, Tokyo Women’s Medical University, 8–1, Kawada-cho, Shinjuku-ku, 162–8666, Tokyo, Japan
| |
Collapse
|