1
|
Weber MC, Clees Z, Buck A, Fischer A, Steffani M, Wilhelm D, Martignoni M, Friess H, Rinkevich Y, Neumann PA. Role of the serosa in intestinal anastomotic healing: insights from in-depth histological analysis of human and murine anastomoses. BJS Open 2024; 8:zrae108. [PMID: 39230923 PMCID: PMC11373408 DOI: 10.1093/bjsopen/zrae108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Anastomotic leakage following colorectal surgery remains a significant complication despite advances in surgical techniques. Recent findings on serosal injury repair in coelomic cavities, such as the peritoneum, challenge the current understanding of the cellular origins and mechanisms underlying intestinal anastomotic healing. Understanding the contribution of each layer of the intestinal wall during anastomotic healing is needed to find new therapeutic strategies to prevent anastomotic leakage. The aim of this experimental study was to investigate the role of the serosal layer of the intestinal wall in anastomotic healing. MATERIALS AND METHODS Comprehensive histologic analysis of human and murine anastomoses was performed to elucidate histologic changes in the different intestinal layers during anastomotic healing. In vivo staining of the extracellular matrix (ECM) in the serosal layer was performed using a fluorophore-conjugated N-hydroxysuccinimide-ester before anastomosis surgery in a murine model. RESULTS Histological examination of both human and murine anastomoses revealed that closure of the serosal layer occurred first during the healing process. In vivo serosal ECM staining demonstrated that a significant portion of the newly formed ECM within the anastomosis was indeed deposited onto the serosal layer. Furthermore, mesenchymal cells within the anastomotic scar were positive for mesothelial cell markers, podoplanin and Wilms tumour protein. CONCLUSIONS In this experimental study, the results suggest that serosal scar formation is an important mechanism for anastomotic integrity in intestinal anastomoses. Mesothelial cells may significantly contribute to scar formation during anastomotic healing through epithelial-to-mesenchymal transition, potentially suggesting a novel therapeutic target to prevent anastomotic leakage by enhancing physiological healing processes.
Collapse
Affiliation(s)
- Marie-Christin Weber
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Zoé Clees
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Annalisa Buck
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Munich, Germany
| | - Adrian Fischer
- Institute of Regenerative Biology and Medicine, Helmholtz Munich, Munich, Germany
| | - Marcella Steffani
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Dirk Wilhelm
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Marc Martignoni
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Munich, Munich, Germany
| | - Philipp-Alexander Neumann
- Department of Surgery, Technical University of Munich, TUM School of Medicine and Health, Munich, Germany
- Institute for Advanced Study, Technical University of Munich, Munich, Germany
| |
Collapse
|
2
|
Szebeni B, Veres-Székely A, Pap D, Bokrossy P, Varga Z, Gaál A, Mihály J, Pállinger É, Takács IM, Pajtók C, Bernáth M, Reusz GS, Szabó AJ, Vannay Á. Extracellular Vesicles of Patients on Peritoneal Dialysis Inhibit the TGF-β- and PDGF-B-Mediated Fibrotic Processes. Cells 2024; 13:605. [PMID: 38607044 PMCID: PMC11011990 DOI: 10.3390/cells13070605] [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: 02/14/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Among patients on peritoneal dialysis (PD), 50-80% will develop peritoneal fibrosis, and 0.5-4.4% will develop life-threatening encapsulating peritoneal sclerosis (EPS). Here, we investigated the role of extracellular vesicles (EVs) on the TGF-β- and PDGF-B-driven processes of peritoneal fibrosis. EVs were isolated from the peritoneal dialysis effluent (PDE) of children receiving continuous ambulatory PD. The impact of PDE-EVs on the epithelial-mesenchymal transition (EMT) and collagen production of the peritoneal mesothelial cells and fibroblasts were investigated in vitro and in vivo in the chlorhexidine digluconate (CG)-induced mice model of peritoneal fibrosis. PDE-EVs showed spherical morphology in the 100 nm size range, and their spectral features, CD63, and annexin positivity were characteristic of EVs. PDE-EVs penetrated into the peritoneal mesothelial cells and fibroblasts and reduced their PDE- or PDGF-B-induced proliferation. Furthermore, PDE-EVs inhibited the PDE- or TGF-β-induced EMT and collagen production of the investigated cell types. PDE-EVs contributed to the mesothelial layer integrity and decreased the submesothelial thickening of CG-treated mice. We demonstrated that PDE-EVs significantly inhibit the PDGF-B- or TGF-β-induced fibrotic processes in vitro and in vivo, suggesting that EVs may contribute to new therapeutic strategies to treat peritoneal fibrosis and other fibroproliferative diseases.
Collapse
Affiliation(s)
- Beáta Szebeni
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Apor Veres-Székely
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Domonkos Pap
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Péter Bokrossy
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
| | - Zoltán Varga
- TTK Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Anikó Gaál
- TTK Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Judith Mihály
- TTK Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, 1117 Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 1089 Budapest, Hungary
| | - István M. Takács
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
| | - Csenge Pajtók
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
| | - Mária Bernáth
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
| | - György S. Reusz
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
| | - Attila J. Szabó
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| | - Ádám Vannay
- Pediatric Center, MTA Center of Excellence, Semmelweis University, 1083 Budapest, Hungary
- HUN-REN–SU Pediatrics and Nephrology Research Group, 1052 Budapest, Hungary
| |
Collapse
|
3
|
Tian Q, Ruan J, Wang Y, Xiao Y, Cheng Q, Chen Y, Li M, Chang K, Yi X. Extracellular succinate derived from ectopic milieu drives adhesion and implantation growth of ectopic endometrial stromal cells via the SUCNR1 signal in endometriosis. Cell Commun Signal 2024; 22:82. [PMID: 38291428 PMCID: PMC10826047 DOI: 10.1186/s12964-023-01415-7] [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: 08/28/2023] [Accepted: 12/02/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND As a dual-function metabolite, succinate has emerged in cell function and plays a key signaling role in linking mitochondrial function to other cellular functions. Succinate accumulation in the cytoplasm is commonly associated with hypoxia in the microenvironment and immune cell activation. Extracellular succinate released into the microenvironment is considered an inflammatory alarm that can be sensed by its membrane receptor SUCNR1, which boosts proinflammatory responses and acts akin to classical hormones and cytokines. Succinate plays an important role in the development of inflammatory diseases. Whether succinate facilitates the progression of endometriosis (EMs), characterized by chronic inflammation and peritoneal adhesion, is worth exploring. OBJECTIVE We mimicked the ectopic milieu in vitro and in vivo to evaluate the main source and potential role of succinate in endometriosis. We assessed the molecular and functional effects of succinate on macrophages and peritoneal mesothelial cells in peritoneal cavity. The effect of succinate/SUCNR1 signaling on ectopic endometrial stromal cells (ESCs) was further explored in this study. METHODS In this study, we used targeted organic acid metabolomics analysis and in vitro assays to assess the potential accumulation of succinate in the peritoneal fluid of EMs patients. We examined its correlation with disease severity, Visual Analogue Scale, and the Endometriosis Fertility Index. Flow cytometry, enzyme linked immunosorbent assay, western blot assay, quantitative real-time PCR, and other molecular biology techniques were used to explore the potential mechanisms. RESULTS By mimicking the ectopic milieu, we constructed an in vitro co-culture system and found that M1 polarized macrophages and that the peritoneal mesothelial cell line (HMrSV5) mainly released succinate into their microenvironment and activated the succinate receptor (SUCNR1) signal, which further polarized the macrophages and significantly enhanced the invasive survival of ESCs, and the adhesion to the peritoneum. We further investigated the pathological effects of extracellular succinate in vivo using a xenograft mouse models of endometriosis. CONCLUSIONS Succinate-SUCNR1 signaling facilitates the creation of inflammatory cells and plays a vital role in EMs progression and peritoneal adhesion. Our work on the molecular mechanisms underlying succinate accumulation and function will help elucidate the phenotypic mysteries of pain and infertility in EMs. Video Abstract.
Collapse
Affiliation(s)
- Qi Tian
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Jingyao Ruan
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yuning Wang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yinping Xiao
- Department of Pathology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Qi Cheng
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Yun Chen
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China
| | - Mingqing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Kaikai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| | - Xiaofang Yi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 419# Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.
| |
Collapse
|
4
|
Uno K, Iyoshi S, Yoshihara M, Kitami K, Mogi K, Fujimoto H, Sugiyama M, Koya Y, Yamakita Y, Nawa A, Kanayama T, Tomita H, Enomoto A, Kajiyama H. Metastatic Voyage of Ovarian Cancer Cells in Ascites with the Assistance of Various Cellular Components. Int J Mol Sci 2022; 23:4383. [PMID: 35457198 PMCID: PMC9031612 DOI: 10.3390/ijms23084383] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and has a unique metastatic route using ascites, known as the transcoelomic root. However, studies on ascites and contained cellular components have not yet been sufficiently clarified. In this review, we focus on the significance of accumulating ascites, contained EOC cells in the form of spheroids, and interaction with non-malignant host cells. To become resistant against anoikis, EOC cells form spheroids in ascites, where epithelial-to-mesenchymal transition stimulated by transforming growth factor-β can be a key pathway. As spheroids form, EOC cells are also gaining the ability to attach and invade the peritoneum to induce intraperitoneal metastasis, as well as resistance to conventional chemotherapy. Recently, accumulating evidence suggests that EOC spheroids in ascites are composed of not only cancer cells, but also non-malignant cells existing with higher abundance than EOC cells in ascites, including macrophages, mesothelial cells, and lymphocytes. Moreover, hetero-cellular spheroids are demonstrated to form more aggregated spheroids and have higher adhesion ability for the mesothelial layer. To improve the poor prognosis, we need to elucidate the mechanisms of spheroid formation and interactions with non-malignant cells in ascites that are a unique tumor microenvironment for EOC.
Collapse
Affiliation(s)
- Kaname Uno
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 223-62 Lund, Sweden
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Discipline of Obstetrics and Gynecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide 5005, Australia
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Yoshihiko Yamakita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (M.S.); (Y.K.); (A.N.)
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (T.K.); (H.T.)
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan;
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466-8560, Japan; (K.U.); (S.I.); (K.K.); (K.M.); (H.F.); (Y.Y.); (H.K.)
| |
Collapse
|
5
|
Peritoneal Restoration by Repurposing Vitamin D Inhibits Ovarian Cancer Dissemination via Blockade of the TGF-β1/Thrombospondin-1 Axis. Matrix Biol 2022; 109:70-90. [PMID: 35339636 DOI: 10.1016/j.matbio.2022.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 02/09/2022] [Accepted: 03/20/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Ovarian cancer (OvCa), a lethal gynecological malignancy, disseminates to the peritoneum. Mesothelial cells (MCs) act as barriers in the abdominal cavity, preventing the adhesion of cancer cells. However, in patients with OvCa, they are transformed into cancer-associated mesothelial cells (CAMs) via mesenchymal transition and form a favorable microenvironment for tumors to promote metastasis. However, attempts for restoring CAMs to their original state have been limited. Here, we investigated whether inhibition of mesenchymal transition and restoration of MCs by vitamin D suppressed the OvCa dissemination in vitro and in vivo. METHODS The effect of vitamin D on the mutual association of MCs and OvCa cells was evaluated using in vitro coculture models and in vivo using a xenograft model. RESULTS Vitamin D restored the CAMs, and thrombospondin-1 (component of the extracellular matrix that is clinically associated with poor prognosis and is highly expressed in peritoneally metastasized OvCa) was found to promote OvCa cell adhesion and proliferation. Mechanistically, TGF-β1 secreted from OvCa cells enhanced thrombospondin-1 expression in CAMs via Smad-dependent TGF-β signaling. Vitamin D inhibited mesenchymal transition in MCs and suppressed thrombospondin-1 expression via vitamin D receptor/Smad3 competition, contributing to the marked reduction in peritoneal dissemination in vivo. Importantly, vitamin D restored CAMs from a stabilized mesenchymal state to the epithelial state and normalized thrombospondin-1 expression in preclinical models that mimic cancerous peritonitis in vivo. CONCLUSIONS MCs are key players in OvCa dissemination and peritoneal restoration and normalization of thrombospondin-1 expression by vitamin D may be a novel strategy for preventing OvCa dissemination.
Collapse
|
6
|
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: 27] [Impact Index Per Article: 9.0] [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
|
7
|
Mullen M, Noia H, Fuh K. Culturing Primary Human Mesothelial Cells. Methods Mol Biol 2022; 2424:147-154. [PMID: 34918292 DOI: 10.1007/978-1-0716-1956-8_9] [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] [Indexed: 06/14/2023]
Abstract
Mesothelial cells line the serosal cavities and associated organs. In order to metastasize to distant organs, ovarian tumor cells must first attach and then clear the mesothelial cells. Therefore, human primary mesothelial cells (HPMCs) are necessary to effectively study ovarian cancer metastases. Here, we describe methods to obtain HPMCs from human omentum and to culture in vitro.
Collapse
Affiliation(s)
- Mary Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hollie Noia
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine Fuh
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine and Alvin J. Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
8
|
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
|
9
|
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: 20] [Impact Index Per Article: 5.0] [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
|
10
|
Post-Surgical Peritoneal Scarring and Key Molecular Mechanisms. Biomolecules 2021; 11:biom11050692. [PMID: 34063089 PMCID: PMC8147932 DOI: 10.3390/biom11050692] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Post-surgical adhesions are internal scar tissue and a major health and economic burden. Adhesions affect and involve the peritoneal lining of the abdominal cavity, which consists of a continuous mesothelial covering of the cavity wall and majority of internal organs. Our understanding of the full pathophysiology of adhesion formation is limited by the fact that the mechanisms regulating normal serosal repair and regeneration of the mesothelial layer are still being elucidated. Emerging evidence suggests that mesothelial cells do not simply form a passive barrier but perform a wide range of important regulatory functions including maintaining a healthy peritoneal homeostasis as well as orchestrating events leading to normal repair or pathological outcomes following injury. Here, we summarise recent advances in our understanding of serosal repair and adhesion formation with an emphasis on molecular mechanisms and novel gene expression signatures associated with these processes. We discuss changes in mesothelial biomolecular marker expression during peritoneal development, which may help, in part, to explain findings in adults from lineage tracing studies using experimental adhesion models. Lastly, we highlight examples of where local tissue specialisation may determine a particular response of peritoneal cells to injury.
Collapse
|
11
|
Shentu Y, Li Y, Xie S, Jiang H, Sun S, Lin R, Chen C, Bai Y, Zhang Y, Zheng C, Zhou Y. Empagliflozin, a sodium glucose cotransporter-2 inhibitor, ameliorates peritoneal fibrosis via suppressing TGF-β/Smad signaling. Int Immunopharmacol 2021; 93:107374. [PMID: 33517222 DOI: 10.1016/j.intimp.2021.107374] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/21/2020] [Accepted: 01/05/2021] [Indexed: 01/26/2023]
Abstract
Sodium glucose cotransporter-2 (SGLT-2) inhibitor has been reported to exert a glucose-lowering effect in the peritoneum exposed to peritoneal dialysis solution. However, whether SGLT-2 inhibitors can regulate peritoneal fibrosis by suppressing TGF-β/Smad signaling is unclear. We aimed to (i) examine the effect of the SGLT-2 inhibitor empagliflozin in reducing inflammatory reaction and preventing peritoneal dialysis solution-induced peritoneal fibrosis and (ii) elucidate the underlying mechanisms. High-glucose peritoneal dialysis solution or transforming growth factor β1 (TGF-β1) was used to induce peritoneal fibrosis in vivo, in a mouse peritoneal dialysis model (C57BL/6 mice) and in human peritoneal mesothelial cells in vitro, to stimulate extracellular matrix accumulation. The effects of empagliflozin and adeno-associated virus-RNAi, which is used to suppress SGLT-2 activity, on peritoneal fibrosis and extracellular matrix were evaluated. The mice that received chronic peritoneal dialysis solution infusions showed typical features of peritoneal fibrosis, including markedly increased peritoneal thickness, excessive matrix deposition, increased peritoneal permeability, and upregulated α-smooth muscle actin and collagen I expression. Empagliflozin treatment or downregulation of SGLT-2 expression significantly ameliorated these pathological changes. Inflammatory cytokines (TNF-α, IL-1β, IL-6) and TGF-β/Smad signaling-associated proteins, such as TGF-β1 and phosphorylated Smad (p-Smad3), decreased in the empagliflozin-treated and SGLT-2 downregulated groups. In addition, empagliflozin treatment and downregulation of SGLT-2 expression reduced the levels of inflammatory cytokines (TNF-α, IL-1β, IL-6), TGF-β1, α-smooth muscle actin, collagen I, and p-Smad3 accumulation in human peritoneal mesothelial cells. Collectively, these results indicated that empagliflozin exerted a clear protective effect on high-glucose peritoneal dialysis-induced peritoneal fibrosis via suppressing TGF-β/Smad signaling.
Collapse
Affiliation(s)
- Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuyang Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shicheng Xie
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Huanchang Jiang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shicheng Sun
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Rixu Lin
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chaosheng Chen
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Institute of Kidney Health, Center for Health Assessment, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chenfei Zheng
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Ying Zhou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
12
|
Mogi K, Yoshihara M, Iyoshi S, Kitami K, Uno K, Tano S, Koya Y, Sugiyama M, Yamakita Y, Nawa A, Tomita H, Kajiyama H. Ovarian Cancer-Associated Mesothelial Cells: Transdifferentiation to Minions of Cancer and Orchestrate Developing Peritoneal Dissemination. Cancers (Basel) 2021; 13:1352. [PMID: 33802781 PMCID: PMC8002484 DOI: 10.3390/cancers13061352] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/18/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer has one of the poorest prognoses among carcinomas. Advanced ovarian cancer often develops ascites and peritoneal dissemination, which is one of the poor prognostic factors. From the perspective of the "seed and soil" hypothesis, the intra-abdominal environment is like the soil for the growth of ovarian cancer (OvCa) and mesothelial cells (MCs) line the top layer of this soil. In recent years, various functions of MCs have been reported, including supporting cancer in the OvCa microenvironment. We refer to OvCa-associated MCs (OCAMs) as MCs that are stimulated by OvCa and contribute to its progression. OCAMs promote OvCa cell adhesion to the peritoneum, invasion, and metastasis. Elucidation of these functions may lead to the identification of novel therapeutic targets that can delay OvCa progression, which is difficult to cure.
Collapse
Affiliation(s)
- Kazumasa Mogi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstr. 19A, 79104 Freiburg, Germany
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
| | - Kaname Uno
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
- Division of Clinical Genetics, Lund University, Sölvegatan 19, 22184 Lund, Sweden
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (Y.K.); (M.S.); (Y.Y.); (A.N.)
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (Y.K.); (M.S.); (Y.Y.); (A.N.)
| | - Yoshihiko Yamakita
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (Y.K.); (M.S.); (Y.Y.); (A.N.)
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (Y.K.); (M.S.); (Y.Y.); (A.N.)
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan;
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8560, Japan; (K.M.); (S.I.); (K.K.); (K.U.); (S.T.)
| |
Collapse
|
13
|
Wang Q, Huang Y, Zhou R, Wu K, Li W, Shi L, Xia Z, Tao K, Wang G, Wang G. Regulation and function of IL-22 in peritoneal adhesion formation after abdominal surgery. Wound Repair Regen 2020; 28:105-117. [PMID: 31148320 DOI: 10.1111/wrr.12740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 05/10/2019] [Accepted: 05/20/2019] [Indexed: 12/20/2022]
Abstract
Peritoneal adhesion occurs frequently after gastrointestinal/gynecological surgery. Tissue repair and regeneration are very important during this process. IL-22 is an important cytokine that is secreted from immune cells but functions on mesenchymal cells, such as mesothelial cells. The objective of this study was to investigate the roles of IL-22 and its regulators during adhesion formation. Postsurgical peritoneal drainage fluid from patients and rodent models was examined by enzyme-linked immunosorbent assay and fluorescence-activated cell sorting. It was observed that IL-22 expression in the abdominal cavity was rapidly induced 12 hours after surgery and then slowly decreased to a lower, steady level for up to 7 days after surgery. However, neutralizing IL-22 at the time point at which the highest level of expression was observed failed to reduce adhesion, but neutralizing IL-22 at a later time point, i.e., 3 days after surgery, prevented adhesion significantly. The IL-22 receptor was induced on the mesothelial membrane, and IL-22BP, an inhibitor of IL-22, was reduced 3 days after surgery. Furthermore, IFN-γ was identified to have the ability to induce IL-22R, and IL-18, which was induced by the infiltrating macrophages, was found to inhibit IL-22BP expression both in vivo and in vitro. Together, these data suggest that IL-22 may promote adhesion formation and that the regulation of IL-22, IL-22R, and IL-22BP may have therapeutic potential to prevent adhesion formation after surgery without disturbing the normal immune process.
Collapse
Affiliation(s)
- Qingbo Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongming Huang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zefeng Xia
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Geng Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
14
|
Monitoring method for transgene expression in target tissue by blood sampling. ACTA ACUST UNITED AC 2019; 24:e00401. [PMID: 31788440 PMCID: PMC6880019 DOI: 10.1016/j.btre.2019.e00401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 01/06/2023]
Abstract
Transgene expressions of simultaneously-administered two plasmid DNA in muscle correlated each other. Transgene expressions of secretable luciferase in muscle and plasma also correlated each other. It was possible to monitor transgene expression in tissues by blood sampling.
In this study, we have developed a novel method to monitor transgene expression in tissues by blood sampling. We administered plasmid DNA (pDNA) encoding non-secretory form of firefly luciferase as a reporter gene and pDNA encoding secretable Gaussia princeps luciferase as a monitor gene simultaneously into mice. Good positive correlations were found between log-transgene expression of the reporter gene and the monitor gene in the treated muscle, between the monitor gene in the treated muscle and plasma, and consequently between the reporter gene in the treated muscle and the monitor gene in plasma after naked pDNA transfer into the muscle of mice. Such positive correlations were also found with gastric serosal surface instillation of naked pDNA, intravenous injection of lipoplex, and hydrodynamics-based injection of naked pDNA. We developed monitoring method of transgene expression in tissues by blood sampling, which was named ‘Therapeutic transgene monitoring (TTM)’, after ‘Therapeutic drug monitoring (TDM)’.
Collapse
|
15
|
Kao HH, Kuo CY, Chen KS, Chen JP. Preparation of Gelatin and Gelatin/Hyaluronic Acid Cryogel Scaffolds for the 3D Culture of Mesothelial Cells and Mesothelium Tissue Regeneration. Int J Mol Sci 2019; 20:4527. [PMID: 31547444 PMCID: PMC6770111 DOI: 10.3390/ijms20184527] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/30/2019] [Accepted: 09/07/2019] [Indexed: 12/14/2022] Open
Abstract
Mesothelial cells are specific epithelial cells that are lined in the serosal cavity and internal organs. Nonetheless, few studies have explored the possibility to culture mesothelial cells in a three-dimensional (3D) scaffold for tissue engineering applications. Towards this end, we fabricated macroporous scaffolds from gelatin and gelatin/hyaluronic acid (HA) by cryogelation, and elucidated the influence of HA on cryogel properties and the cellular phenotype of mesothelial cells cultured within the 3D scaffolds. The incorporation of HA was found not to significantly change the pore size, porosity, water uptake kinetics, and swelling ratios of the cryogel scaffolds, but led to a faster scaffold degradation in the collagenase solution. Adding 5% HA in the composite cryogels also decreased the ultimate compressive stress (strain) and toughness of the scaffold, but enhanced the elastic modulus. From the in vitro cell culture, rat mesothelial cells showed quantitative cell viability in gelatin (G) and gelatin/HA (GH) cryogels. Nonetheless, mesothelial cells cultured in GH cryogels showed a change in the cell morphology and cytoskeleton arrangement, reduced cell proliferation rate, and downregulation of the mesothelium specific maker gene expression. The production of key mesothelium proteins E-cadherin and calretinin were also reduced in the GH cryogels. Choosing the best G cryogels for in vivo studies, the cell/cryogel construct was used for the transplantation of allograft mesothelial cells for mesothelium reconstruction in rats. A mesothelium layer similar to the native mesothelium tissue could be obtained 21 days post-implantation, based on hematoxylin and eosin (H&E) and immunohistochemical staining.
Collapse
Affiliation(s)
- Hao-Hsi Kao
- Division of Nephrology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Kuo-Su Chen
- Division of Nephrology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
- School of Medicine, Chang Gung University, Kwei-San, Taoyuan 33303, Taiwan.
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan.
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan.
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
16
|
Namvar S, Woolf AS, Zeef LA, Wilm T, Wilm B, Herrick SE. Functional molecules in mesothelial-to-mesenchymal transition revealed by transcriptome analyses. J Pathol 2018; 245:491-501. [PMID: 29774544 PMCID: PMC6055603 DOI: 10.1002/path.5101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 03/01/2018] [Accepted: 05/12/2018] [Indexed: 12/13/2022]
Abstract
Peritoneal fibrosis is a common complication of abdominal and pelvic surgery, and can also be triggered by peritoneal dialysis, resulting in treatment failure. In these settings, fibrosis is driven by activated myofibroblasts that are considered to be partly derived by mesothelial‐to‐mesenchymal transition (MMT). We hypothesized that, if the molecular signature of MMT could be better defined, these insights could be exploited to block this pathological cellular transition. Rat peritoneal mesothelial cells were purified by the use of an antibody against HBME1, a protein present on mesothelial cell microvilli, and streptavidin nanobead technology. After exposure of sorted cells to a well‐known mediator of MMT, transforming growth factor (TGF)‐β1, RNA sequencing was undertaken to define the transcriptomes of mesothelial cells before and during early‐phase MMT. MMT was associated with dysregulation of transcripts encoding molecules involved in insulin‐like growth factor (IGF) and bone morphogenetic protein (BMP) signalling. The application of either recombinant BMP4 or IGF‐binding protein 4 (IGFBP4) ameliorated TGF‐β1‐induced MMT in culture, as judged from the retention of epithelial morphological and molecular phenotypes, and reduced migration. Furthermore, peritoneal tissue from peritoneal dialysis patients showed less prominent immunostaining than control tissue for IGFBP4 and BMP4 on the peritoneal surface. In a mouse model of TGF‐β1‐induced peritoneal thickening, BMP4 immunostaining on the peritoneal surface was attenuated as compared with healthy controls. Finally, genetic lineage tracing of mesothelial cells was used in mice with peritoneal injury. In this model, administration of BMP4 ameliorated the injury‐induced shape change and migration of mesothelial cells. Our findings demonstrate a distinctive MMT signature, and highlight the therapeutic potential for BMP4, and possibly IGFBP4, to reduce MMT. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sara Namvar
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Leo Ah Zeef
- The Bioinformatics Core Facility, The University of Manchester, Manchester, UK
| | - Thomas Wilm
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Bettina Wilm
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Sarah E Herrick
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
17
|
Rossi L, Battistelli C, de Turris V, Noce V, Zwergel C, Valente S, Moioli A, Manzione A, Palladino M, Bordoni V, Domenici A, Menè P, Mai A, Tripodi M, Strippoli R. HDAC1 inhibition by MS-275 in mesothelial cells limits cellular invasion and promotes MMT reversal. Sci Rep 2018; 8:8492. [PMID: 29855565 PMCID: PMC5981641 DOI: 10.1038/s41598-018-26319-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
Peritoneal fibrosis is a pathological alteration of the peritoneal membrane occurring in a variety of conditions including peritoneal dialysis (PD), post-surgery adhesions and peritoneal metastases. The acquisition of invasive and pro-fibrotic abilities by mesothelial cells (MCs) through induction of MMT, a cell-specific form of EMT, plays a main role in this process. Aim of this study was to evaluate possible effects of histone deacetylase (HDAC) inhibitors, key components of the epigenetic machinery, in counteracting MMT observed in MCs isolated from effluent of PD patients. HDAC inhibitors with different class/isoform selectivity have been used for pharmacological inhibition. While the effect of other inhibitors was limited to a partial E-cadherin re-expression, MS-275, a HDAC1-3 inhibitor, promoted: (i) downregulation of mesenchymal markers (MMP2, Col1A1, PAI-1, TGFβ1, TGFβRI) (ii) upregulation of epithelial markers (E-cadherin, Occludin), (iii) reacquisition of an epithelial-like morphology and (iv) marked reduction of cellular invasiveness. Results were confirmed by HDAC1 genetic silencing. Mechanistically, MS-275 causes: (i) increase of nuclear histone H3 acetylation (ii) rescue of the acetylation profile on E-cadherin promoter, (iii) Snail functional impairment. Overall, our study, pinpointing a role for HDAC1, revealed a new player in the regulation of peritoneal fibrosis, providing the rationale for future therapeutic opportunities.
Collapse
Affiliation(s)
- Lucia Rossi
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Valeria de Turris
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Valeria Noce
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Clemens Zwergel
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Alessandra Moioli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Andrea Manzione
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Marco Palladino
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Veronica Bordoni
- Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy
| | - Alessandro Domenici
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Paolo Menè
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Marco Tripodi
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy. .,Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy.
| | - Raffaele Strippoli
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy. .,Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy.
| |
Collapse
|
18
|
González-Tortuero E, Rodríguez-Beltrán J, Radek R, Blázquez J, Rodríguez-Rojas A. Clay-induced DNA breaks as a path for genetic diversity, antibiotic resistance, and asbestos carcinogenesis. Sci Rep 2018; 8:8504. [PMID: 29855603 PMCID: PMC5981458 DOI: 10.1038/s41598-018-26958-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/23/2018] [Indexed: 11/09/2022] Open
Abstract
Natural clays and synthetic nanofibres can have a severe impact on human health. After several decades of research, the molecular mechanism of how asbestos induces cancer is not well understood. Different fibres, including asbestos, can penetrate cell membranes and introduce foreign DNA in bacterial and eukaryotic cells. Incubating Escherichia coli under friction forces with sepiolite, a clayey material, or with asbestos, causes double-strand DNA breaks. Antibiotics and clays are used together in animal husbandry, the mutagenic effect of these fibres could be a pathway to antibiotic resistance due to the friction provided by peristalsis of the gut from farm animals in addition to horizontal gene transfer. Moreover, we raise the possibility that the same mechanism could generate bacteria diversity in natural scenarios, playing a role in the evolution of species. Finally, we provide a new model on how asbestos may promote mutagenesis and cancer based on the observed mechanical genotoxicity.
Collapse
Affiliation(s)
- Enrique González-Tortuero
- Department of Ecosystem Research, Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Müggelseedamm 301, 12587, Berlin, Germany.,Berlin Centre for Genomics in Biodiversity Research (BeGenDiv), Königin-Luise-Straße 6-8, 14195, Berlin, Germany.,Institute for Genome Sciences, University of Maryland Baltimore School of Medicine, 670 West Baltimore Street, 21201, Baltimore, MD, USA
| | - Jerónimo Rodríguez-Beltrán
- Department of Microbial Biotechnology, Spanish National Center for Biotechnology, Calle Darwin 3, 28049, Madrid, Spain
| | - Renate Radek
- Evolutionary Biology, Institut für Biologie, Freie Universität Berlin, Berlin, Germany
| | - Jesús Blázquez
- Department of Microbial Biotechnology, Spanish National Center for Biotechnology, Calle Darwin 3, 28049, Madrid, Spain
| | | |
Collapse
|
19
|
Kienzle A, Servais AB, Ysasi AB, Gibney BC, Valenzuela CD, Wagner WL, Ackermann M, Mentzer SJ. Free-Floating Mesothelial Cells in Pleural Fluid After Lung Surgery. Front Med (Lausanne) 2018; 5:89. [PMID: 29675416 PMCID: PMC5895720 DOI: 10.3389/fmed.2018.00089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/22/2018] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVES The mesothelium, the surface layer of the heart, lung, bowel, liver, and tunica vaginalis, is a complex tissue implicated in organ-specific diseases and regenerative biology; however, the mechanism of mesothelial repair after surgical injury is unknown. Previous observations indicated seeding of denuded mesothelium by free-floating mesothelial cells may contribute to mesothelial healing. In this study, we investigated the prevalence of mesothelial cells in pleural fluid during the 7 days following pulmonary surgery. STUDY DESIGN Flow cytometry was employed to study pleural fluid of 45 patients after lung resection or transplantation. We used histologically validated mesothelial markers (CD71 and WT1) to estimate the prevalence of mesothelial cells. RESULTS The viability of pleural fluid cells approached 100%. Leukocytes and mesothelial cells were identified in the pleural fluid within the first week after surgery. The leukocyte concentration was relatively stable at all time points. In contrast, mesothelial cells, identified by CD71 and WT1 peaked on POD3. The broad expression of CD71 molecule in postoperative pleural fluid suggests that many of the free-floating non-leukocyte cells were activated or proliferative mesothelial cells. CONCLUSION We demonstrated that pleural fluid post lung surgery is a source of mesothelial cells; most of these cells appear to be viable and, as shown by CD71 staining, activated mesothelial cells. The observed peak of mesothelial cells on POD3 is consistent with a potential reparative role of free-floating mesothelial cells after pulmonary surgery.
Collapse
Affiliation(s)
- Arne Kienzle
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrew B. Servais
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Alexandra B. Ysasi
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Barry C. Gibney
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Cristian D. Valenzuela
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Willi L. Wagner
- Department of Diagnostic and Interventional Radiology, Translational Lung Research Center Heidelberg (TLRC), Member of German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Steven J. Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
Rehrauer H, Wu L, Blum W, Pecze L, Henzi T, Serre-Beinier V, Aquino C, Vrugt B, de Perrot M, Schwaller B, Felley-Bosco E. How asbestos drives the tissue towards tumors: YAP activation, macrophage and mesothelial precursor recruitment, RNA editing, and somatic mutations. Oncogene 2018; 37:2645-2659. [PMID: 29507420 PMCID: PMC5955862 DOI: 10.1038/s41388-018-0153-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/11/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022]
Abstract
Chronic exposure to intraperitoneal asbestos triggered a marked response in the mesothelium well before tumor development. Macrophages, mesothelial precursor cells, cytokines, and growth factors accumulated in the peritoneal lavage. Transcriptome profiling revealed YAP/TAZ activation in inflamed mesothelium with further activation in tumors, paralleled by increased levels of cells with nuclear YAP/TAZ. Arg1 was one of the highest upregulated genes in inflamed tissue and tumor. Inflamed tissue showed increased levels of single-nucleotide variations, with an RNA-editing signature, which were even higher in the tumor samples. Subcutaneous injection of asbestos-treated, but tumor-free mice with syngeneic mesothelioma tumor cells resulted in a significantly higher incidence of tumor growth when compared to naïve mice supporting the role of the environment in tumor progression.
Collapse
Affiliation(s)
- Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Walter Blum
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Lazslo Pecze
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Thomas Henzi
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | | | - Catherine Aquino
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8057, Zurich, Switzerland
| | - Bart Vrugt
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital University Health Network, University of Toronto, Toronto, ON, Canada
| | - Beat Schwaller
- Department of Medicine, Unit of Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700, Fribourg, Switzerland
| | - Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, Lungen- und Thoraxonkologie Zentrum, University Hospital Zürich, Sternwartstrasse 14, 8091, Zurich, Switzerland.
| |
Collapse
|
21
|
Hekking LH, van den Born J. Feasibility of Mesothelial Transplantation during Experimental Peritoneal Dialysis and Peritonitis. Int J Artif Organs 2018; 30:513-9. [PMID: 17628852 DOI: 10.1177/039139880703000609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The mesothelial cell layer lining the peritoneum orchestrates peritoneal homeostasis. Continuous exposure to peritoneal dialysis fluids and episodes of peritonitis may damage the monolayer irreversibly, eventually leading to adhesion formation and fibrosis/sclerosis of the peritoneum. Autologous mesothelial cell transplantation is thought to be one of the options to reduce dysfunction of the peritoneal membrane. In this article we will review the mesothelial cell transplantation experiments performed in the field of peritoneal dialysis and peritonitis. In addition we will focus on the trouble shooting using cultured autologous mesothelial cells for transplantation.
Collapse
Affiliation(s)
- L H Hekking
- Department of Cellular Architecture and Dynamics, University of Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
22
|
Mutsaers SE, Prêle CM, Lansley SM, Herrick SE. The Origin of Regenerating Mesothelium: A Historical Perspective. Int J Artif Organs 2018; 30:484-94. [PMID: 17628849 DOI: 10.1177/039139880703000606] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bichat first described the mesothelium in 1827 but despite its early discovery, it has only been in recent years that its importance both in health and disease has been realised. One area still poorly understood is that of the mechanisms regulating mesothelial repair. Mesothelial cells are derived from the mesoderm but express many epithelial characteristics. However, mesothelium does not heal in the same way as other epithelial-like cells. Epithelium heals by centripetal migration, with cells at the edge of the wound proliferating and migrating into the injured area. Hertzler in 1919 noted that both large and small peritoneal injuries healed within the same time frame, concluding that the mesothelium could not heal solely by centripetal migration. The exact mechanisms involved in mesothelial regeneration following injury are controversial with a number of proposals suggested to explain the origin of the regenerating cells. This review will examine these proposals and give some insights into the likely mechanisms involved.
Collapse
Affiliation(s)
- S E Mutsaers
- Anatomical Pathology, PathWest Laboratory Medicine WA, Nedlands 6009, WA, Australia.
| | | | | | | |
Collapse
|
23
|
Gotloib L, Gotloib LC, Khrizman V. The use of Peritoneal Mesothelium as a Potential Source of Adult Stem Cells. Int J Artif Organs 2018; 30:501-12. [PMID: 17628851 DOI: 10.1177/039139880703000608] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
At the dawn of the 21st century, classical curative medicine is being challenged by the fact that efforts to fight and prevent not a few diseases, are in many circumstances, beyond the power of the pharmacological armamentarium of the medical profession. On the other hand, replacement of lost function by mechanical or biophysical devices, or even by organ transplantation, prolongs life but generally derives in new and, at times, unsolvable problems. Regenerative therapy using stem cells began a revolutionary trend that may well change both the therapeutic approach to not a few of the diseases resulting from failing organs, as well as the fate and quality of life of millions of patients. The presence of pluripotent mesenchymal cells in the mesothelial monolayer as well as in the submesothelial connective tissue raises the possibility of using the peritoneal mesothelium in regenerative therapies. This perception of the problem is also based on observations made in humans as well as in laboratory animals showing bone, bone marrow, cartilaginous tissue, glomerular-like structures and creation of blood conducts, pathological situations (mesothelioma, sclerosing peritonitis), or after in vivo or ex vivo experimental interventions. The main concept emerging from this information is that peritoneal mesothelial cells are endowed with such a degree of plasticity that, if placed in the appropriate micro-environment, they have a remarkable potential to generate other mesenchymal-derived cell lines. Intensive research is required to define the best environmental conditions to take advantage of this plasticity and make the peritoneal mesothelium an actual option to be applied in regenerative medicine.
Collapse
Affiliation(s)
- L Gotloib
- Laboratory for Experimental Nephrology, Ha'Emek Medical Center, Afula 18101, Israel.
| | | | | |
Collapse
|
24
|
Di Paolo N, Sacchi G, Del Vecchio MT, Nicolai GA, Brardi S, Garosi G. State of the Art on Autologous Mesothelial Transplant in Animals and Humans. Int J Artif Organs 2018; 30:456-76. [PMID: 17628847 DOI: 10.1177/039139880703000604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sixteen years ago rabbit and human mesothelial cells were successsfully cultured and autoimplanted. The aim of the study was merely to demostrate that mesothelial implant was possible and interesting not only in peritoneal dialysis, but also in the vaster field of medicine and surgery concerning all the mesothelial districts of the body. The aim of this paper is to recollect the steps which have led to autolougous mesothelial transplantation and verify if the tecnique has been validated and adopted by others. Review of the literature published in the last 15 years shows that intraperitoneal transplantation of mesothelial cells has been effective in reducing the formation of peritoneal adhesions, and in remodeling the area of mesothelial denudation. New studies on the mesothelial cell opened the way to costruction of transplantable tissue-engineered artificial peritoneum, to the utilization of mesothelial progenitor cells and to find simple metods to collect autologous mesothelial cells. Finally mesothelial trasnsplantation may represent a new neovascular therapy in the prevention and treatment of ischemic coronaric heart disease.
Collapse
Affiliation(s)
- N Di Paolo
- Department of Nephrology, Dialysis and Kidney Transplant, Siena University Hospital, Siena, Italy.
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Adhesions are bands of fibrous tissue that form between opposing organs and the peritoneum, restricting vital intrapleural and abdominal movement. They remain a major problem in abdominal surgery, occurring in more than three fourths of patients following laparotomy. Adhesions result when injury to the mesothelium is not repaired by mesothelial cells and can be viewed as scar tissue formation. The mechanism of mesothelial healing suggested the involvement of stem cells in the process. It has long been known that peritoneal wounds heal in the same amount of time regardless of size. Therefore, the mesothelium could not regenerate solely by proliferation and centripetal migration of cells at the wound edge as occurs in the healing of skin epithelium. Several studies suggest the presence of i) mesothelial stem cells that can differentiate into mesothelial cells and a few other phenotypes and/or ii) that mesothelial cells are themselves stem cells. Other studies have suggested that adult stem cells in the muscle underlying the peritoneum can differentiate into mesothelial cells and contribute to healing. Prevention of abdominal adhesions have been accomplished by delivery of autologous mesothelial cells and multipotent adult stem cells isolated from skeletal muscle. Adult stem cells from sources other than the serosal tissue offer an alternative treatment modality to prevent the formation of abdominal adhesions.
Collapse
Affiliation(s)
- P A Lucas
- Department of Orthopaedic Surgery, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
26
|
Herrick SE, Mutsaers SE. The Potential of Mesothelial Cells in Tissue Engineering and Regenerative Medicine Applications. Int J Artif Organs 2018; 30:527-40. [PMID: 17628854 DOI: 10.1177/039139880703000611] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Injury to the serosa through injurious agents such as radiation, surgery, infection and disease results in the loss of the protective surface mesothelium and often leads to fibrous adhesion formation. Mechanisms that increase the rate of mesothialisation are therefore actively being investigated in order to reduce the formation of adhesions. These include intraperitoneal delivery of cultured mesothelial cells as well as administration of factors that are known to increase mesothelial proliferation and migration. An exciting alternative that has only recently received attention, is the possible role of mesothelial progenitor cells in the repair and regeneration of denuded serosal areas. Accumulating evidence suggests that such a population exists and under certain conditions is able to form a number of defined cell types indicating a degree of plasticity. Such properties may explain the extensive use of mesothelial cells in various tissue engineering applications including the development of vascular conduits and peripheral nerve replacements. It is likely that with the rapid explosion in the fields of tissue engineering and regenerative medicine, a greater understanding of the potential of mesothelial progenitor cells to repair, replace and possibly regenerate damaged or defective tissue will be uncovered.
Collapse
Affiliation(s)
- S E Herrick
- School of Medicine, Faculty of Medical and Human Sciences, University of Manchester, Oxford Road, Manchester, UK.
| | | |
Collapse
|
27
|
Injectable thermosensitive hydrogel containing hyaluronic acid and chitosan as a barrier for prevention of postoperative peritoneal adhesion. Carbohydr Polym 2017; 173:721-731. [PMID: 28732919 DOI: 10.1016/j.carbpol.2017.06.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/16/2017] [Accepted: 06/05/2017] [Indexed: 01/28/2023]
Abstract
Peritoneal adhesion is one of the common complications after abdominal surgery. Injectable thermosensitive hydrogel could serve as an ideal barrier to prevent this postoperative tissue adhesion. In this study, poly(N-isopropylacrylamide) (PNIPAm) was grafted to chitosan (CS) and the polymer was further conjugated with hyaluronic acid (HA) to form thermosensitive HA-CS-PNIPAm hydrogel. Aqueous solutions of PNIPAm and HA-CS-PNIPAm at 10%(w/v) are both free-flowing and injectable at room temperature and exhibit sol-gel phase transition around 31°C; however, HA-CS-PNIPAm shows less volume shrinkage after gelation and higher complex modulus than PNIPAm. Cell culture studies indicate both injectable hydrogel show barrier effects to reduce fibroblasts penetration while induce little cytotoxicity in vitro. From a sidewall defect-bowel abrasion model in rats, significant reduction of postoperative peritoneal adhesion was found for peritoneal defects treated with HA-CS-PNIPAm compared with those treated with PNIPAm and untreated controls from gross and histological evaluation. Furthermore, HA-CS-PNIPAm did not interfere with normal peritoneal tissue healing and did not elicit acute toxicity from blood analysis and tissue biopsy examination. By taking advantage of the easy handling and placement properties of HA-CS-PNIPAm during application, this copolymer hydrogel would be a potentially ideal injectable anti-adhesion barrier after abdominal surgeries.
Collapse
|
28
|
Sanchez-Martin D, Uldrick TS, Kwak H, Ohnuki H, Polizzotto MN, Annunziata CM, Raffeld M, Wyvill KM, Aleman K, Wang V, Marshall VA, Whitby D, Yarchoan R, Tosato G. Evidence for a Mesothelial Origin of Body Cavity Effusion Lymphomas. J Natl Cancer Inst 2017; 109:3078996. [PMID: 28376153 DOI: 10.1093/jnci/djx016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/20/2017] [Indexed: 12/19/2022] Open
Abstract
Background Primary effusion lymphoma (PEL) is a Kaposi's sarcoma herpes virus (KSHV)-induced lymphoma that typically arises in body cavities of HIV-infected patients. PEL cells are often co-infected with Epstein-Barr virus (EBV). "PEL-like" lymphoma is a KSHV-unrelated lymphoma that arises in body cavities of HIV-negative patients. "PEL-like" lymphoma is sometimes EBV positive. The derivation of PEL/"PEL-like" cells is unclear. Methods Mesothelial cells were cultured from body cavity effusions of 23 patients. Cell proliferation, cytokine secretion, marker phenotypes, KSHV/EBV infection, and clonality were evaluated by standard methods. Gene expression was measured by quantitative polymerase chain reaction and immunoblotting. A mouse model of PEL (3 mice/group) was used to evaluate tumorigenicity. Results We found that the mesothelia derived from six effusions of HIV-infected patients with PEL or other KSHV-associated diseases contained rare KSHV + or EBV + mesothelial cells. After extended culture (16-17 weeks), some mesothelial cells underwent a trans-differentiation process, generating lymphoid-type CD45 + /B220 + , CD5 + , CD27 + , CD43 + , CD11c + , and CD3 - cells resembling "B1-cells," most commonly found in mouse body cavities. These "B1-like" cells were short lived. However, long-term KSHV + EBV - and EBV + KSHV - clonal cell lines emerged from mesothelial cultures from two patients that were clonally distinct from the monoclonal or polyclonal B-cell populations found in the patients' original effusions. Conclusions Mesothelial-to-lymphoid transformation is a newly identified in vitro process that generates "B1-like" cells and is associated with the emergence of long-lived KSHV or EBV-infected cell lines in KSHV-infected patients. These results identify mesothelial cultures as a source of PEL cells and lymphoid cells in humans.
Collapse
Affiliation(s)
- David Sanchez-Martin
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas S Uldrick
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Hyeongil Kwak
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Hidetaka Ohnuki
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mark N Polizzotto
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mark Raffeld
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Kathleen M Wyvill
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Karen Aleman
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Victoria Wang
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Vickie A Marshall
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Denise Whitby
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Robert Yarchoan
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
29
|
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: 93] [Impact Index Per Article: 11.6] [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
|
30
|
Lansley SM, Cheah HM, Lee YCG. Role of MCP-1 in pleural effusion development in a carrageenan-induced murine model of pleurisy. Respirology 2016; 22:758-763. [PMID: 27878909 DOI: 10.1111/resp.12951] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/12/2016] [Accepted: 09/03/2016] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND OBJECTIVE Exudative pleural effusions affect over 1500 patients per million population each year. The pathobiology of pleural exudate formation remains unclear. Our recent study revealed monocyte chemotactic protein-1 (MCP-1) as a key driver of fibrinolytic-induced exudate effusion while another study found a role for MCP-1 in malignant effusion formation. In the present study, we further evaluated the role of MCP-1 in the development of pleural effusion in a mouse model of acute pleural inflammation. METHODS λ-Carrageenan (CAR) was injected into the pleural cavity of CD1 mice and pleural effusion volume measured up to 16 h post-injection. Pleural effusion and serum protein and MCP-1 concentrations were measured and differential cell counts performed in fluids. Mice were also treated with either intraperitoneal (i) anti-MCP-1 antibody or isotype control or (ii) an MCP-1 receptor (CCR2) antagonist or vehicle control 12 h prior to and at the time of CAR injection. RESULTS Intrapleural CAR induced significant pleural fluid accumulation (300.0 ± 49.9 μL) in mice after 4 h. Pleural fluid MCP-1 concentrations were significantly higher than corresponding serum MCP-1 (144 603 ± 23 204 pg/mL vs 3703 ± 801 pg/mL, P < 0.0001). A significant decrease in pleural fluid formation was seen both with anti-MCP-1 antibody (median (interquartile range, IQR): 36 (0-168) μL vs controls 290 (70-436) μL; P = 0.02) or CCR2 antagonist (153 (30-222) μL vs controls 240 (151-331) μL, P = 0.0049). CONCLUSIONS Blockade of MCP-1 activity significantly reduced inflammatory pleural effusion formation in a CAR model. Together with recent successes in MCP-1 blockade in other effusion formation models, our data strongly support clinical evaluation of MCP-1 antagonists as a novel approach to pleural fluid management.
Collapse
Affiliation(s)
- Sally M Lansley
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Hui Min Cheah
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Y C Gary Lee
- Centre for Respiratory Health, School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia.,Respiratory Department, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
31
|
Cheah HM, Lansley SM, Varano Della Vergiliana JF, Tan AL, Thomas R, Leong SL, Creaney J, Lee YCG. Malignant pleural fluid from mesothelioma has potent biological activities. Respirology 2016; 22:192-199. [PMID: 27560254 DOI: 10.1111/resp.12874] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/30/2016] [Accepted: 06/13/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Malignant pleural effusion (MPE) affects >90% of mesothelioma patients. Research on MPE has focused on its physical impact on breathlessness; MPE is rich in growth mediators but its contribution to tumour biology has not been investigated. We aimed to examine the potential effects of MPE in promoting growth, migration and chemo-resistance of mesothelioma. METHODS Pleural fluid samples from 151 patients (56 mesothelioma, 60 metastatic pleural cancer and 35 benign) were used. Seven validated human mesothelioma cell lines and three primary cultured mesothelioma lines were employed. RESULTS Pleural fluid from mesothelioma patients (diluted to 30%) consistently stimulated cell proliferation (trypan-blue cell viability assay) in five mesothelioma cell lines tested by (median) 2.23-fold over controls (all P < 0.0001). The fluid also induced cell migration by (median) 2.13-fold in six mesothelioma cell lines using scratch-wound assay. In a murine flank model of mesothelioma, tumour infused with daily instillations of pleural fluid grew significantly faster over saline controls (median 52.5 cm2 vs 28.0 cm2 at day 13, P = 0.028). Addition of MPE (diluted to 30%) to culture media significantly protected mesothelioma from cisplatin/pemetrexed-induced cell death in all three cell lines tested (median fold reduction of 1.29, 1.98 and 3.90, all P < 0.001 vs control). The growth effects of matched pleural fluid and cultured mesothelioma cells from the same patients did not differ significantly from unmatched pairs. CONCLUSION This 'proof-of-concept' study reveals potent biological capabilities of malignant pleural fluid in mesothelioma pathobiology.
Collapse
Affiliation(s)
- Hui Min Cheah
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia.,School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Sally M Lansley
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia
| | | | - Ai Ling Tan
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia.,School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Rajesh Thomas
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia.,School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia.,Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Su Lyn Leong
- School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia.,Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Jenette Creaney
- School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Yun Chor Gary Lee
- Pleural Medicine Unit, Institute for Respiratory Health, Perth, Western Australia, Australia.,School of Medicine & Pharmacology, University of Western Australia, Perth, Western Australia, Australia.,Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
32
|
Dauleh S, Santeramo I, Fielding C, Ward K, Herrmann A, Murray P, Wilm B. Characterisation of Cultured Mesothelial Cells Derived from the Murine Adult Omentum. PLoS One 2016; 11:e0158997. [PMID: 27403660 PMCID: PMC4942062 DOI: 10.1371/journal.pone.0158997] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/24/2016] [Indexed: 12/16/2022] Open
Abstract
The human omentum has been long regarded as a healing patch, used by surgeons for its ability to immunomodulate, repair and vascularise injured tissues. A major component of the omentum are mesothelial cells, which display some of the characteristics of mesenchymal stem/stromal cells. For instance, lineage tracing studies have shown that mesothelial cells give rise to adipocytes and vascular smooth muscle cells, and human and rat mesothelial cells have been shown to differentiate into osteoblast- and adipocyte-like cells in vitro, indicating that they have considerable plasticity. However, so far, long-term cultures of mesothelial cells have not been successfully established due to early senescence. Here, we demonstrate that mesothelial cells isolated from the mouse omentum could be cultured for more than 30 passages. While epithelial markers were downregulated over passages in the mesothelial cells, their mesenchymal profile remained unchanged. Early passage mesothelial cells displayed clonogenicitiy, expressed several stem cell markers, and up to passage 5 and 13, respectively, could differentiate along the adipogenic and osteogenic lineages, demonstrating stem/progenitor characteristics and differentiation potential.
Collapse
Affiliation(s)
- Sumaya Dauleh
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Ilaria Santeramo
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Claire Fielding
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kelly Ward
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Anne Herrmann
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Kitayama J, Yamaguchi H, Ishigami H, Matsuzaki K, Sata N. Intraperitoneal Mesenchymal Cells Promote the Development of Peritoneal Metastasis Partly by Supporting Long Migration of Disseminated Tumor Cells. PLoS One 2016; 11:e0154542. [PMID: 27136922 PMCID: PMC4854412 DOI: 10.1371/journal.pone.0154542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/14/2016] [Indexed: 11/18/2022] Open
Abstract
The human peritoneal cavity contains a small number of free cells of mesenchymal cell lineage. Intraperitoneal mesenchymal cells (PMC) play supportive roles in metastasis formation on the peritoneum. In this study, we found that PMC, when co-cultuerd with human gastric cancer cells, MKN45, enhanced the proliferation of MKN45 when cultured at low, but not high, cellular density. Also, PMC suppressed apoptotic cell death of MKN45 only under low density culture conditions. Time-lapse videoanalysis clearly demonstrated that PMC randomly migrated more vigorously than did MKN45, and strongly enhanced the migration behavior of co-cultured MKN45. In fact, the majority of MKN45 migrated together in direct physical contact with PMC, and the sum of migration lengths from original position of co-cultured MKN45 for 48 hours was approximately 10 times longer than that of MKN45 cultured alone. Our data suggest that enhanced migration can increase the chance of direct contact or positional proximity among sparcely distributed MKN45, which may bring survival advantages to tumor cells. This may be one of the important mechanisms of peritoneal metastasis, since only a small number of tumor cells are considered to be disseminated in the early step of metastasis formation on the peritoneum.
Collapse
Affiliation(s)
- Joji Kitayama
- Department of Gastrointestinal Surgery, Jichi Medical University, Tochigi, Japan
- * E-mail:
| | - Hironori Yamaguchi
- Department of Gastrointestinal Surgery, Jichi Medical University, Tochigi, Japan
| | | | | | - Naohiro Sata
- Department of Gastrointestinal Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
34
|
Molecular Mechanisms Underlying Peritoneal EMT and Fibrosis. Stem Cells Int 2016; 2016:3543678. [PMID: 26941801 PMCID: PMC4752998 DOI: 10.1155/2016/3543678] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/10/2016] [Indexed: 12/26/2022] Open
Abstract
Peritoneal dialysis is a form of renal replacement alternative to the hemodialysis. During this treatment, the peritoneal membrane acts as a permeable barrier for exchange of solutes and water. Continual exposure to dialysis solutions, as well as episodes of peritonitis and hemoperitoneum, can cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy, eventually leading to discontinuation of the peritoneal dialysis. Among the different events controlling this pathological process, epithelial to mesenchymal transition of mesothelial cells plays a main role in the induction of fibrosis and in subsequent functional deterioration of the peritoneal membrane. Here, the main extracellular inducers and cellular players are described. Moreover, signaling pathways acting during this process are elucidated, with emphasis on signals delivered by TGF-β family members and by Toll-like/IL-1β receptors. The understanding of 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
|
35
|
Lansley SM, Cheah HM, Varano Della Vergiliana JF, Chakera A, Lee YCG. Tissue plasminogen activator potently stimulates pleural effusion via a monocyte chemotactic protein-1-dependent mechanism. Am J Respir Cell Mol Biol 2015; 53:105-12. [PMID: 25474480 DOI: 10.1165/rcmb.2014-0017oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pleural infection is common. Evacuation of infected pleural fluid is essential for successful treatment, but it is often difficult because of adhesions/loculations within the effusion and the viscosity of the fluid. Intrapleural delivery of tissue plasminogen activator (tPA) (to break the adhesions) and deoxyribonuclease (DNase) (to reduce fluid viscosity) has recently been shown to improve clinical outcomes in a large randomized study of pleural infection. Clinical studies of intrapleural fibrinolytic therapy have consistently shown subsequent production of large effusions, the mechanism(s) of which are unknown. We aimed to determine the mechanism by which tPA induces exudative fluid formation. Intrapleural tPA, with or without DNase, significantly induced pleural fluid accumulation in CD1 mice (tPA alone: median [interquartile range], 53.5 [30-355] μl) compared with DNase alone or vehicle controls (both, 0.0 [0.0-0.0] μl) after 6 hours. Fluid induction was reproduced after intrapleural delivery of streptokinase and urokinase, indicating a class effect. Pleural fluid monocyte chemotactic protein (MCP)-1 levels strongly correlated with effusion volume (r = 0.7302; P = 0.003), and were significantly higher than MCP-1 levels in corresponding sera. Mice treated with anti-MCP-1 antibody (P < 0.0001) or MCP-1 receptor antagonist (P = 0.0049) demonstrated a significant decrease in tPA-induced pleural fluid formation (by up to 85%). Our data implicate MCP-1 as the key molecule governing tPA-induced fluid accumulation. The role of MCP-1 in the development of other exudative effusions warrants examination.
Collapse
Affiliation(s)
- Sally M Lansley
- 1 Pleural Disease Unit, Lung Institute of Western Australia, Perth, Western Australia, Australia
| | - Hui Min Cheah
- 1 Pleural Disease Unit, Lung Institute of Western Australia, Perth, Western Australia, Australia.,2 School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and
| | | | - Aron Chakera
- 2 School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and.,Departments of 3 Renal Medicine and
| | - Y C Gary Lee
- 1 Pleural Disease Unit, Lung Institute of Western Australia, Perth, Western Australia, Australia.,2 School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia; and.,4 Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
36
|
Lachaud CC, Rodriguez-Campins B, Hmadcha A, Soria B. Use of Mesothelial Cells and Biological Matrices for Tissue Engineering of Simple Epithelium Surrogates. Front Bioeng Biotechnol 2015; 3:117. [PMID: 26347862 PMCID: PMC4538307 DOI: 10.3389/fbioe.2015.00117] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022] Open
Abstract
Tissue-engineering technologies have progressed rapidly through last decades resulting in the manufacture of quite complex bioartificial tissues with potential use for human organ and tissue regeneration. The manufacture of avascular monolayered tissues such as simple squamous epithelia was initiated a few decades ago and is attracting increasing interest. Their relative morphostructural simplicity makes of their biomimetization a goal, which is currently accessible. The mesothelium is a simple squamous epithelium in nature and is the monolayered tissue lining the walls of large celomic cavities (peritoneal, pericardial, and pleural) and internal organs housed inside. Interestingly, mesothelial cells can be harvested in clinically relevant numbers from several anatomical sources and not less important, they also display high transdifferentiation capacities and are low immunogenic characteristics, which endow these cells with therapeutic interest. Their combination with a suitable scaffold (biocompatible, degradable, and non-immunogenic) may allow the manufacture of tailored serosal membranes biomimetics with potential spanning a wide range of therapeutic applications, principally for the regeneration of simple squamous-like epithelia such as the visceral and parietal mesothelium vascular endothelium and corneal endothelium among others. Herein, we review recent research progresses in mesothelial cells biology and their clinical sources. We make a particular emphasis on reviewing the different types of biological scaffolds suitable for the manufacture of serosal mesothelial membranes biomimetics. Finally, we also review progresses made in mesothelial cells-based therapeutic applications and propose some possible future directions.
Collapse
Affiliation(s)
- Christian Claude Lachaud
- Andalusian Center for Molecular Biology and Regenerative Medicine - Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER) , Seville , Spain ; Centro de Investigación en Red sobre Diabetes y Enfermedades Metabólicas (CIBERDEM) , Madrid , Spain
| | - Berta Rodriguez-Campins
- Departamento de I+D, New Biotechnic S.A. , Seville , Spain ; Fundación Andaluza de Investigación y Desarrollo (FAID) , Seville , Spain
| | - Abdelkrim Hmadcha
- Andalusian Center for Molecular Biology and Regenerative Medicine - Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER) , Seville , Spain ; Centro de Investigación en Red sobre Diabetes y Enfermedades Metabólicas (CIBERDEM) , Madrid , Spain
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine - Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER) , Seville , Spain ; Centro de Investigación en Red sobre Diabetes y Enfermedades Metabólicas (CIBERDEM) , Madrid , Spain
| |
Collapse
|
37
|
Rapid reperitonealization and wound healing in a preclinical model of abdominal trauma repair with a composite mesh. Int J Surg 2015; 22:86-91. [PMID: 26166739 DOI: 10.1016/j.ijsu.2015.06.071] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/18/2015] [Indexed: 11/23/2022]
Abstract
PURPOSE Peritoneal tissue healing is characterized by the simultaneous repopulation of mesothelial cells and the formation of neoperitoneum. Despite the common use of mesh products for abdominal wall repair, there are few investigations of how these materials may impact the peritoneal healing process. Here, we utilized an animal model of abdominal trauma to specifically investigate the peritoneal healing process in conjunction with a composite (poliglecaprone 25-coated polypropylene) mesh. METHODS Abdominal wall injury was simulated in New Zealand White rabbits and peritoneal tissue was covered with composite mesh and fixed with peripheral sutures. Animals were sacrificed at regular intervals (up to 28 days) for macroscopic and microscopic evaluation. RESULTS Mesothelial cells were consistently identified on the surface of the central areas of the implanted mesh as early as 3-5 days after implantation. From day 7 onward, the entire mesh surface was covered by neoperitoneum which matured over the remaining study intervals. Fibroblast ingrowth of the mesh was apparent by day 5 and increased over time, concurrent with fragmentation of the film on the composite mesh. CONCLUSIONS These results suggest that composite mesh products used for abdominal wall repair do not significantly delay mesothelial repopulation. Study results also support the hypothesis that mesothelial cells involved in healing are derived, at least in part in this model, from free-floating precursor cells located within the peritoneal cavity.
Collapse
|
38
|
Mutsaers SE, Birnie K, Lansley S, Herrick SE, Lim CB, Prêle CM. Mesothelial cells in tissue repair and fibrosis. Front Pharmacol 2015; 6:113. [PMID: 26106328 PMCID: PMC4460327 DOI: 10.3389/fphar.2015.00113] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/12/2015] [Indexed: 12/21/2022] Open
Abstract
Mesothelial cells are fundamental to the maintenance of serosal integrity and homeostasis and play a critical role in normal serosal repair following injury. However, when normal repair mechanisms breakdown, mesothelial cells take on a profibrotic role, secreting inflammatory, and profibrotic mediators, differentiating and migrating into the injured tissues where they contribute to fibrogenesis. The development of new molecular and cell tracking techniques has made it possible to examine the origin of fibrotic cells within damaged tissues and to elucidate the roles they play in inflammation and fibrosis. In addition to secreting proinflammatory mediators and contributing to both coagulation and fibrinolysis, mesothelial cells undergo mesothelial-to-mesenchymal transition, a process analogous to epithelial-to-mesenchymal transition, and become fibrogenic cells. Fibrogenic mesothelial cells have now been identified in tissues where they have not previously been thought to occur, such as within the parenchyma of the fibrotic lung. These findings show a direct role for mesothelial cells in fibrogenesis and open therapeutic strategies to prevent or reverse the fibrotic process.
Collapse
Affiliation(s)
- Steven E Mutsaers
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Kimberly Birnie
- Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Sally Lansley
- Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Sarah E Herrick
- Institute of Inflammation and Repair, Faculty of Medical and Human Sciences and Manchester Academic Health Science Centre, University of Manchester , Manchester, UK
| | - Chuan-Bian Lim
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| | - Cecilia M Prêle
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia and Harry Perkins Institute of Medical Research , Nedlands, WA, Australia ; Institute for Respiratory Health, Centre for Asthma, Allergy and Respiratory Research, School of Medicine and Pharmacology, University of Western Australia , Nedlands, WA, Australia
| |
Collapse
|
39
|
Kitayama J, Emoto S, Yamaguchi H, Ishigami H, Yamashita H, Seto Y, Matsuzaki K, Watanabe T. CD90(+)CD45(-) intraperitoneal mesothelial-like cells inhibit T cell activation by production of arginase I. Cell Immunol 2014; 288:8-14. [PMID: 24556645 DOI: 10.1016/j.cellimm.2014.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 12/13/2013] [Accepted: 01/24/2014] [Indexed: 12/17/2022]
Abstract
In this study, we analyzed intraperitoneal cells recovered from human samples and found that CD90(+)CD45(-) cells exist as a minor population but vigorously grow in culture, showing the morphological features of mesothelial cells (MC). Interestingly, the MC highly expressed arginase I and markedly suppressed T cell proliferation with the reduction of CD3 ζ chain expression in T cells stimulated by coated anti-CD3 mAb. The addition of nor-NOHA (500 μM), or L-arginine (1 mM) mostly restored the inhibitory effect of MC on T cell proliferation as well as the reduced expression of CD3 ζ chain. The expression level of CD3 ζ chain in T cells in the peritoneal cavity was significantly down-regulated from circulating T cells. These results suggest that intraperitoneal free MC have immunomodulatory functions through the control of L-arginine level, and thus may play significant roles in the pathogenesis of various diseases in the peritoneal cavity.
Collapse
Affiliation(s)
- Joji Kitayama
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan.
| | - Shigenobu Emoto
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
| | | | | | | | - Yasuyuki Seto
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
40
|
Kitayama J, Emoto S, Yamaguchi H, Ishigami H, Watanabe T. CD90+ mesothelial-like cells in peritoneal fluid promote peritoneal metastasis by forming a tumor permissive microenvironment. PLoS One 2014; 9:e86516. [PMID: 24466130 PMCID: PMC3897715 DOI: 10.1371/journal.pone.0086516] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023] Open
Abstract
The peritoneal cavity is a common target of metastatic gastrointestinal and ovarian cancer cells, but the mechanisms leading to peritoneal metastasis have not been fully elucidated. In this study, we examined the roles of cells in peritoneal fluids on the development of peritoneal metastasis. We found that a minor subset of human intraperitoneal cells with CD90(+)/CD45(−) phenotype vigorously grew in culture with mesothelial-like appearance. The mesothelial-like cells (MLC) displayed the characteristics of mesenchymal stem cell, such as differentiating into adipocytes, osteocytes, and chondrocytes, and suppressing T cell proliferation. These cells highly expressed type I collagen, vimentin, α-smooth muscle actin and fibroblast activated protein-α by the stimulation with TGF-β, which is characteristic of activated myofibroblasts. Intraperitoneal co-injection of MLCs with the human gastric cancer cell line, MKN45, significantly enhanced the rate of metastatic formation in the peritoneum of nude mice. Histological examination revealed that many MLCs were engrafted in metastatic nodules and were mainly located at the fibrous area. Dasatinib, a potent tyrosine kinase inhibitor, strongly inhibited the proliferation of MLCs but not MKN45 in vitro. Nevertheless, oral administration of Dasatinib significantly inhibited the development of peritoneal metastasis of MKN45, and resulted in reduced fibrillar formation of metastatic nodules. These results suggest floating MLCs in the peritoneal fluids support the development of peritoneal metastasis possibly through the production of the permissive microenvironment, and thus the functional blockade of MLCs is a reasonable strategy to treat recurrent abdominal malignancies.
Collapse
Affiliation(s)
- Joji Kitayama
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
- * E-mail:
| | - Shigenobu Emoto
- Department of Surgical Oncology, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
41
|
Kanda R, Hamada C, Kaneko K, Nakano T, Wakabayashi K, Hara K, Io H, Horikoshi S, Tomino Y. Paracrine effects of transplanted mesothelial cells isolated from temperature-sensitive SV40 large T-antigen gene transgenic rats during peritoneal repair. Nephrol Dial Transplant 2013; 29:289-300. [PMID: 24081857 PMCID: PMC3910341 DOI: 10.1093/ndt/gft397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The prevention and restoration of peritoneal damage is a critical mission in peritoneal dialysis (PD). Transplantation of mesothelial cells has been suggested to suppress peritoneal injury during PD. Few studies have examined the efficacy and safety of cell transplantation. We evaluated the paracrine effects of mesothelial transplantation during peritoneal repair using immortalized temperature-sensitive mesothelial cells (TSMCs) in chlorhexidine gluconate (CG)-induced peritoneal fibrosis rats. METHODS Continuous-infusion pumps containing 8% CG were placed into the abdominal cavity for 21 days. After the removal of the pumps, the TSMCs were injected into the peritoneal cavity at Day 22 (Tx-1 group) or 29 (Tx-2 group). Morphological findings and mRNA expressions of regeneration-related factors were examined at Days 22, 29 and 35. RESULTS Peritoneal thickness was aggravated in the Tx-1 group. Levels of transforming growth factor (TGF)-β, vascular endothelial growth factor (VEGF) and matrix metalloproteinase-2 mRNA in the Tx-1 group at Day 35 were comparable with those at Day 22. The levels of Snail, B-Raf and ERK-1, markers of epithelial to mesenchymal transition and of the RAS/MAPK pathway in the Tx-1 group, were significantly higher than those in the Tx-2 group. TGF-β and VEGF were produced from the transplanted mesothelial cells and the surrounding cells in the Tx-1 group. CONCLUSION It appears that the paracrine effect of transplanted mesothelial cells during peritoneal repair is associated with its surrounding condition. It is important to determine the most appropriate time for developing peritoneal repair through mesothelial transplantation.
Collapse
Affiliation(s)
- Reo Kanda
- Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Christodoulidis G, Tsilioni I, Spyridakis ME, Kiropoulos T, Oikonomidi S, Koukoulis G, Tepetes K. Matrix metaloproteinase-2 and -9 serum levels as potential markers of intraperitoneal adhesions. J INVEST SURG 2013; 26:134-140. [PMID: 23514055 DOI: 10.3109/08941939.2012.730599] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the value of matrix metalloproteinases-2 (MMP-2) and -9 (MMP-9) as prognostic serum markers for intraperitoneal adhesions. BACKGROUND Postoperative adhesions are associated with serious complications responsible for increased patient's morbidity. METHODS Forty-eight rabbits were used and randomized into groups A, B, C, and D. Abdominal laparotomy and experimental adhesion formation model was carried out. In group A, 60 mL of N/S 0.9% were instilled intraperitoneally, in group B 60 mL of icodextrin 4% were instilled intraperitoneally, in group C 0.1 mL/kg of dimetindene maleate were administered intravenously, and in group D both agents were administered. Prior to euthanasia 0.5 mL of blood was obtained. The type, the surface area of adhesions, and serum concentration of MMPs were assessed. RESULTS The mean surface area and Zuhlke classification of adhesions of groups B, C, and D has been proved to be significantly lower compared to group A. Serum MMP-2 levels were significantly higher in groups B and D than in group A, while group D was higher when compared to group C. Serum MMP-9 levels were significantly higher in group D compared to groups A, B, and C. Serum MMP-9 was the most accurate test to differentiate between animals with and without adhesions with a sensitivity of 81.8% and a specificity of 100% at a cut-off point of 21.5 (AUC = 0.934). CONCLUSIONS The administration of icodextrin 4% and dimetindene maleate seems to prevent postoperative adhesion formation. Serum levels of MMP-2 and MMP-9 may serve as prognostic markers to identify postoperative adhesions.
Collapse
|
43
|
Cannata A, Petrella D, Russo CF, Bruschi G, Fratto P, Gambacorta M, Martinelli L. Postsurgical Intrapericardial Adhesions: Mechanisms of Formation and Prevention. Ann Thorac Surg 2013; 95:1818-26. [DOI: 10.1016/j.athoracsur.2012.11.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/07/2012] [Accepted: 11/12/2012] [Indexed: 10/27/2022]
|
44
|
Negrini D, Moriondo A. Pleural function and lymphatics. Acta Physiol (Oxf) 2013; 207:244-59. [PMID: 23009260 DOI: 10.1111/apha.12016] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 07/24/2012] [Accepted: 09/17/2012] [Indexed: 11/26/2022]
Abstract
The pleural space plays an important role in respiratory function as the negative intrapleural pressure regimen ensures lung expansion and in the mean time maintains the tight mechanical coupling between the lung and the chest wall. The efficiency of the lung-chest wall coupling depends upon pleural liquid volume, which in turn reflects the balance between the filtration of fluid into and its egress out of the cavity. While filtration occurs through a single mechanism passively driving fluid from the interstitium of the parietal pleura into the cavity, several mechanisms may co-operate to remove pleural fluid. Among these, the pleural lymphatic system emerges as the most important one in quantitative terms and the only one able to cope with variable pleural fluid volume and drainage requirements. In this review, we present a detailed account of the actual knowledge on: (a) the complex morphology of the pleural lymphatic system, (b) the mechanism supporting pleural lymph formation and propulsion, (c) the dependence of pleural lymphatic function upon local tissue mechanics and (d) the effect of lymphatic inefficiency in the development of clinically severe pleural and, more in general, respiratory pathologies.
Collapse
Affiliation(s)
- D. Negrini
- Department of Surgical and Morphological Sciences; University of Insubria; Varese; Italy
| | - A. Moriondo
- Department of Surgical and Morphological Sciences; University of Insubria; Varese; Italy
| |
Collapse
|
45
|
Metamorphosis of mesothelial cells with active horizontal motility in tissue culture. Sci Rep 2013; 3:1144. [PMID: 23359855 PMCID: PMC3556594 DOI: 10.1038/srep01144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 12/27/2012] [Indexed: 02/07/2023] Open
Abstract
Mesothelial cells, which have diverse roles in physiology and pathology, constitute the mesothelium along with connective tissue and the basement membrane; the mesothelium serves to shield the somatic cavities. After mesothelial injury, mesothelial cells undergo tissue recovery. However, the mechanism of mesothelial regeneration remains poorly understood. In this study, we used confocal time-lapse microscopy to demonstrate that transformed mesothelial cells (MeT5A) and mouse peritoneal mesothelial cells can randomly migrate between cells in cell culture and in ex vivo tissue culture, respectively. Moreover, peritoneal mesothelial cells changed their morphology from a flattened shape to a cuboidal one prior to the migration. Conversely, MDCKII epithelial cells forming tight cell–cell contacts with one another do not alter the arrangement of adjacent cells during movement. Our evidence complements the current hypotheses of mesothelial regeneration and suggests that certain types of differentiated mesothelial cells undergo morphological changes before initiating migration to repair injured sites.
Collapse
|
46
|
Petricevic J, Punda H, Brakus SM, Vukojevic K, Govorko DK, Alfirevic D, Kvesic A, Saraga-Babic M. Immunolocalization of nestin, mesothelin and epithelial membrane antigen (EMA) in developing and adult serous membranes and mesotheliomas. Acta Histochem 2012; 114:469-79. [PMID: 22113177 DOI: 10.1016/j.acthis.2011.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/24/2011] [Accepted: 08/28/2011] [Indexed: 11/17/2022]
Abstract
The spatial and temporal distribution of epithelial membrane antigen (EMA), mesothelin and nestin was immunohistochemically analyzed in developing and adult human serous membranes and mesotheliomas in order to detect possible differences in the course of mesenchymal to epithelial transformation, which is associated with differentiation of mesothelial cells during normal development and tumorigenesis. Pleura and pericardium developing from the visceral mesoderm gradually transform into mesothelial cells and connective tissue. EMA appeared in mesothelium of both serous membranes during the early fetal period, whereas during further development, EMA expression was retained only in the pericardial mesothelium. It increased in both pleural mesothelium and connective tissue. Mesothelin appeared first in pericardial submesothelial cells and later in surface mesothelium, while in pleura it was immediately localized in mesothelium. In adult serous membranes, EMA and mesothelin were predominantly expressed in mesothelium. Nestin never appeared in mesothelium, but in connective tissues and myocardial cells and subsequently decreased during development, apart from in the walls of blood vessels. Mesothelial cells in the two serous membranes developed in two separate developmental pathways. We speculate that submesothelial pericardial and mesothelial pleural cells might belong to a population of stem cells. In epithelioid mesotheliomas, 13% of cells expressed nestin, 39% EMA and 7% mesothelin.
Collapse
Affiliation(s)
- Josko Petricevic
- Department of Pathology, Cytology and Forensic Medicine, University Clinical Hospital Mostar, Bosnia and Herzegovina
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gong C, Yang B, Qian Z, Zhao X, Wu Q, Qi X, Wang Y, Guo G, Kan B, Luo F, Wei Y. Improving intraperitoneal chemotherapeutic effect and preventing postsurgical adhesions simultaneously with biodegradable micelles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8:963-73. [DOI: 10.1016/j.nano.2011.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 08/25/2011] [Accepted: 10/25/2011] [Indexed: 12/13/2022]
|
48
|
FOX SIMONA, LOH SUZANNES, MAHENDRAN SATHISHK, GARLEPP MICHAELJ. Regulated chemokine gene expression in mouse mesothelioma and mesothelial cells: TNF-α upregulates both CC and CXC chemokine genes. Oncol Rep 2012; 28:707-13. [DOI: 10.3892/or.2012.1809] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 03/23/2012] [Indexed: 11/06/2022] Open
|
49
|
Pathophysiological changes to the peritoneal membrane during PD-related peritonitis: the role of mesothelial cells. Mediators Inflamm 2012; 2012:484167. [PMID: 22577250 PMCID: PMC3337720 DOI: 10.1155/2012/484167] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 01/18/2012] [Accepted: 01/18/2012] [Indexed: 01/08/2023] Open
Abstract
The success of peritoneal dialysis (PD) is dependent on the structural and functional integrity of the
peritoneal membrane. The mesothelium lines the peritoneal membrane and is the first line of
defense against chemical and/or bacterial insult. Peritonitis remains a major complication of PD and
is a predominant cause of technique failure, morbidity and mortality amongst PD patients. With
appropriate antibiotic treatment, peritonitis resolves without further complications, but in some PD
patients excessive peritoneal inflammatory responses lead to mesothelial cell exfoliation and
thickening of the submesothelium, resulting in peritoneal fibrosis and sclerosis. The detrimental
changes in the peritoneal membrane structure and function correlate with the number and severity
of peritonitis episodes and the need for catheter removal. There is evidence that despite clinical
resolution of peritonitis, increased levels of inflammatory and fibrotic mediators may persist in the
peritoneal cavity, signifying persistent injury to the mesothelial cells. This review will describe the
structural and functional changes that occur in the peritoneal membrane during peritonitis and how
mesothelial cells contribute to these changes and respond to infection. The latter part of the review
discusses the potential of mesothelial cell transplantation and genetic manipulation in the
preservation of the peritoneal membrane.
Collapse
|
50
|
Pathophysiology of the peritoneal membrane during peritoneal dialysis: the role of hyaluronan. J Biomed Biotechnol 2011; 2011:180594. [PMID: 22203782 PMCID: PMC3238805 DOI: 10.1155/2011/180594] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/11/2011] [Accepted: 09/12/2011] [Indexed: 01/28/2023] Open
Abstract
During peritoneal dialysis (PD), constant exposure of mesothelial cells to bioincompatible PD solutions results in the denudation of the mesothelial monolayer and impairment of mesothelial cell function. Hyaluronan, a major component of extracellular matrices, is synthesized by mesothelial cells and contributes to remesothelialization, maintenance of cell phenotype, and tissue remodeling and provides structural support to the peritoneal membrane. Chronic peritoneal inflammation is observed in long-term PD patients and is associated with increased hyaluronan synthesis. During inflammation, depolymerization of hyaluronan may occur with the generation of hyaluronan fragments. In contrast to native hyaluronan which offers a protective role to the peritoneum, hyaluronan fragments exacerbate inflammatory and fibrotic processes and therefore assist in the destruction of the tissue. This paper will discuss the contribution of mesothelial cells to peritoneal membrane alterations that are induced by PD and the putative role of hyaluronan in these processes.
Collapse
|