1
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Richbourg N, Irakoze N, Peyton SR. Tenascin-C Activation of Lung Fibroblasts in a 3D Synthetic Lung Extracellular Matrix Mimic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301493. [PMID: 37227134 PMCID: PMC10528529 DOI: 10.1002/adma.202301493] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Indexed: 05/26/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
2
|
Fu X, Wang Q, Du H, Hao H. CXCL8 and the peritoneal metastasis of ovarian and gastric cancer. Front Immunol 2023; 14:1159061. [PMID: 37377954 PMCID: PMC10291199 DOI: 10.3389/fimmu.2023.1159061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
CXCL8 is the most representative chemokine produced autocrine or paracrine by tumor cells, endothelial cells and lymphocytes. It can play a key role in normal tissues and tumors by activating PI3K-Akt, PLC, JAK-STAT, and other signaling pathways after combining with CXCR1/2. The incidence of peritoneal metastasis in ovarian and gastric cancer is extremely high. The structure of the peritoneum and various peritoneal-related cells supports the peritoneal metastasis of cancers, which readily produces a poor prognosis, low 5-year survival rate, and the death of patients. Studies show that CXCL8 is excessively secreted in a variety of cancers. Thus, this paper will further elaborate on the mechanism of CXCL8 and the peritoneal metastasis of ovarian and gastric cancer to provide a theoretical basis for the proposal of new methods for the prevention, diagnosis, and treatment of cancer peritoneal metastasis.
Collapse
|
3
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Irakoze N, Richbourg N, Peyton SR. Tenascin-C activation of lung fibroblasts in a 3D synthetic lung extracellular matrix mimic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529926. [PMID: 36865293 PMCID: PMC9980292 DOI: 10.1101/2023.02.24.529926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that contain the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro . Here, we developed a synthetic, bioactive hydrogel that mimics the native lung modulus, and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which promotes quiescence of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C activated these hydrogel-encapsulated HLFs in a manner reflective of their native in vivo responses. We propose this lung hydrogel platform as a tunable, synthetic approach to study the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
4
|
Role of Peritoneal Mesothelial Cells in the Progression of Peritoneal Metastases. Cancers (Basel) 2022; 14:cancers14122856. [PMID: 35740521 PMCID: PMC9221366 DOI: 10.3390/cancers14122856] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Peritoneal metastatic cancer comprises a heterogeneous group of primary tumors that originate in the peritoneal cavity or metastasize into the peritoneal cavity from a different origin. Metastasis is a characteristic of end-stage disease, often indicative of a poor prognosis with limited treatment options. Peritoneal mesothelial cells (PMCs) are a thin layer of cells present on the surface of the peritoneum. They display differentiated characteristics in embryonic development and adults, representing the first cell layer encountering peritoneal tumors to affect their progression. PMCs have been traditionally considered a barrier to the intraperitoneal implantation and metastasis of tumors; however, recent studies indicate that PMCs can either inhibit or actively promote tumor progression through distinct mechanisms. This article presents a review of the role of PMCs in the progression of peritoneum implanted tumors, offering new ideas for therapeutic targets and related research.
Collapse
|
5
|
Wang Z, Liu J, Huang H, Ye M, Li X, Wu R, Liu H, Song Y. Metastasis-associated fibroblasts: an emerging target for metastatic cancer. Biomark Res 2021; 9:47. [PMID: 34112258 PMCID: PMC8194104 DOI: 10.1186/s40364-021-00305-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Metastasis suggests a poor prognosis for cancer patients, and treatment strategies for metastatic cancer are still very limited. Numerous studies have shown that cancer-associated fibroblasts (CAFs), a large component of the tumor microenvironment, contribute to tumor metastasis. Stromal fibroblasts at metastatic sites are different from CAFs within primary tumors and can be termed metastasis-associated fibroblasts (MAFs), and they also make great contributions to the establishment of metastatic lesions and the therapeutic resistance of metastatic tumors. MAFs are capable of remodeling the extracellular matrix of metastatic tumors, modulating immune cells in the tumor microenvironment, promoting angiogenesis and enhancing malignant tumor phenotypes. Thus, MAFs can help establish premetastatic niches and mediate resistance to therapeutic strategies, including immunotherapy and antiangiogenic therapy. The results of preclinical studies suggest that targeting MAFs can alleviate the progression of metastatic cancer and mitigate therapeutic resistance, indicating that MAFs are a promising target for metastatic cancer. Here, we comprehensively summarize the existing evidence on MAFs and discuss their origins, generation, functions and related therapeutic strategies in an effort to provide a better understanding of MAFs and offer treatment perspectives for metastatic cancer.
Collapse
Affiliation(s)
- Zimu Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Jiaxin Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, 210002, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China
| | - Xinying Li
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, Nanjing University School of Medicine, 210008, Nanjing, Jiangsu, China
| | - Ranpu Wu
- Department of Respiratory Medicine, Jinling Hospital, Southeast University of Medicine, 210009, Nanjing, Jiangsu, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, #305, East Zhongshan Road, 210002, Nanjing, Jiangsu, China.
| |
Collapse
|
6
|
Demuytere J, Ceelen W, Van Dorpe J, Hoorens A. The role of the peritoneal microenvironment in the pathogenesis of colorectal peritoneal carcinomatosis. Exp Mol Pathol 2020; 115:104442. [PMID: 32305340 DOI: 10.1016/j.yexmp.2020.104442] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 01/06/2023]
Abstract
Recent insights have implicated mesothelial-to-mesenchymal transition (MMT) as a mechanism by which mesothelial cells can transdifferentiate into cancer-associated fibroblasts (CAFs) in several cancers metastasizing to the peritoneum. However, this was not evaluated extensively in colorectal cancer. We examined the presumed mesothelial origin of CAFs in three types of colorectal carcinoma: conventional type adenocarcinoma, mucinous carcinoma and signet ring cell carcinoma. We evaluated the expression of mesothelial, mesenchymal, angiogenesis and colorectal cancer-related markers in peritoneal samples of twelve colorectal cancer patients with peritoneal carcinomatosis and four control patients by immunohistochemistry. We observed morphological and immunohistochemical changes in the vicinity of tumor implants in all studied colorectal cancer types. Mesothelial cells acquired a spindle-shaped myofibroblast-like morphology, lost expression of mesothelial markers, and gained expression of mesenchymal markers. Analysis of consecutive tissue sections and double staining for mesothelial and mesenchymal markers revealed overlap in expression of mesothelial and CAF markers. These findings are highly suggestive of a mesothelial origin of CAFs in peritoneal carcinomatosis in colorectal cancer. Interfering with the process of MMT might be a valuable approach in treating and preventing peritoneal carcinomatosis. Differences observed between colorectal cancer types suggest that one single strategy might not be applicable.
Collapse
Affiliation(s)
- Jesse Demuytere
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Wim Ceelen
- Laboratory of Experimental Surgery, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, Ghent University Hospital, Ghent, Belgium..
| |
Collapse
|
7
|
Umakoshi M, Takahashi S, Itoh G, Kuriyama S, Sasaki Y, Yanagihara K, Yashiro M, Maeda D, Goto A, Tanaka M. Macrophage-mediated transfer of cancer-derived components to stromal cells contributes to establishment of a pro-tumor microenvironment. Oncogene 2018; 38:2162-2176. [PMID: 30459356 DOI: 10.1038/s41388-018-0564-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 10/01/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022]
Abstract
Tumor-derived extracellular vesicles (TEVs) secreted into the blood create a pre-metastatic niche in distant organs; however, it is unclear how TEVs are delivered and how they affect stromal cells in the tumor microenvironment. Tumor-associated macrophages (TAMs) have pivotal roles in cancer progression by interacting with cancer cells and other stromal cells. Here, we report a novel function of TAMs: delivery and transmission of TEV contents. TEV-incorporating macrophages (TEV-MΦs) showed increased invasiveness and were disseminated widely. Upon contact with host stromal cells (peritoneal mesothelial cells (PMCs), fibroblasts, and endothelial cells), TEV-MΦs released membrane blebs containing TEVs, a process dependent upon localized activation of caspase-3 in MΦs. Scattered blebs were incorporated into stromal cells, leading to transfer of cancer-derived RNA and proteins such as TGF-β, activated Src, Wnt3, and HIF1α. TEV-MΦ-secreted blebs containing cancer-derived components contributed to myofibroblastic changes in recipient stromal cells. TEVs delivered by MΦs penetrated deep into the parenchyma of the stomach in TEV-injected mice, and transmitted TEVs to PMCs lining the stomach surface; this process induced PMCs to undergo mesothelial-mesenchymal transition. PMCs infiltrated the gastric wall and created a niche, thereby promoting tumor invasion. Depletion of MΦs prevented these events. Moreover, TEV-MΦs created a pro-metastatic niche. Taken together, these results suggest a novel function for TAMs: transfer of cancer-derived components to surrounding stromal cells and induction of a pro-tumor microenvironment via an increase in the number of CAF-like cells.
Collapse
Affiliation(s)
- Michinobu Umakoshi
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - So Takahashi
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Gastroenterology and Neurology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Yuto Sasaki
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, 1-1 Tegata Gakuenmachi, Akita, 010-8502, Japan
| | - Kazuyoshi Yanagihara
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwa-shi, Chiba, 277-0882, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8545, Japan
| | - Daichi Maeda
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Clinical Genomics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suitashi, Osaka, 565-0871, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.
| |
Collapse
|
8
|
Mesothelial to mesenchyme transition as a major developmental and pathological player in trunk organs and their cavities. Commun Biol 2018; 1:170. [PMID: 30345394 PMCID: PMC6191446 DOI: 10.1038/s42003-018-0180-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022] Open
Abstract
The internal organs embedded in the cavities are lined by an epithelial monolayer termed the mesothelium. The mesothelium is increasingly implicated in driving various internal organ pathologies, as many of the normal embryonic developmental pathways acting in mesothelial cells, such as those regulating epithelial-to-mesenchymal transition, also drive disease progression in adult life. Here, we summarize observations from different animal models and organ systems that collectively point toward a central role of epithelial-to-mesenchymal transition in driving tissue fibrosis, acute scarring, and cancer metastasis. Thus, drugs targeting pathways of mesothelium’s transition may have broad therapeutic benefits in patients suffering from these diseases. Tim Koopmans and Yuval Rinkevich review recent findings linking the mesothelium’s embryonic programs that drive epithelial-to-mesenchyme transition with adult pathologies, such as fibrosis, acute scarring, and cancer metastasis. They highlight new avenues for drug development that would target pathways of the mesothelium’s mesenchymal transition.
Collapse
|
9
|
Li Q, Li B, Li Q, Wei S, He Z, Huang X, Wang L, Xia Y, Xu Z, Li Z, Wang W, Yang L, Zhang D, Xu Z. Exosomal miR-21-5p derived from gastric cancer promotes peritoneal metastasis via mesothelial-to-mesenchymal transition. Cell Death Dis 2018; 9:854. [PMID: 30154401 PMCID: PMC6113299 DOI: 10.1038/s41419-018-0928-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023]
Abstract
Peritoneal metastasis is a primary metastatic route for gastric cancers, and the mechanisms underlying this process are still unclear. Peritoneal mesothelial cells (PMCs) undergo mesothelial-to-mesenchymal transition (MMT) to provide a favorable environment for metastatic cancer cells. In this study, we investigated how the exosomal miR-21-5p induces MMT and promotes peritoneal metastasis. Gastric cancer (GC)-derived exosomes were identified by transmission electron microscopy and western blot analysis, then the uptake of exosomes was confirmed by PKH-67 staining. The expression of miR-21-5p and SMAD7 were measured by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, and the interactions between miR-21-5p and its target genes SMAD7 were confirmed by Luciferase reporter assays. The MMT of PMCs was determined by invasion assays, adhesion assays, immunofluorescent assay, and western blot. Meanwhile, mouse model of tumor peritoneal dissemination model was performed to investigate the role of exosomal miR-21-5p in peritoneal metastasis in vivo. We found that PMCs could internalize GC-derived exosomal miR-21-5p and led to increased levels of miR-21-5p in PMCs. Through various types of in vitro and in vivo assays, we confirmed that exosomal miR-21-5p was able to induce MMT of PMCs and promote tumor peritoneal metastasis. Moreover, our study revealed that this process was promoted by exosomal miR-21-5p through activating TGF-β/Smad pathway via targeting SMAD7. Altogether, our data suggest that exosomal miR-21-5p induces MMT of PMCs and promote cancer peritoneal dissemination by targeting SMAD7. The exosomal miR-21-5p may be a novel therapeutic target for GC peritoneal metastasis.
Collapse
Affiliation(s)
- Qiang Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Qing Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Song Wei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Xiaoxu Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Yiwen Xia
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zhipeng Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Li Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China.
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu province, China
| |
Collapse
|
10
|
Yazdani S, Bansal R, Prakash J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv Drug Deliv Rev 2017; 121:101-116. [PMID: 28720422 DOI: 10.1016/j.addr.2017.07.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/20/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
Myofibroblasts are the key players in extracellular matrix remodeling, a core phenomenon in numerous devastating fibrotic diseases. Not only in organ fibrosis, but also the pivotal role of myofibroblasts in tumor progression, invasion and metastasis has recently been highlighted. Myofibroblast targeting has gained tremendous attention in order to inhibit the progression of incurable fibrotic diseases, or to limit the myofibroblast-induced tumor progression and metastasis. In this review, we outline the origin of myofibroblasts, their general characteristics and functions during fibrosis progression in three major organs: liver, kidneys and lungs as well as in cancer. We will then discuss the state-of-the art drug targeting technologies to myofibroblasts in context of the above-mentioned organs and tumor microenvironment. The overall objective of this review is therefore to advance our understanding in drug targeting to myofibroblasts, and concurrently identify opportunities and challenges for designing new strategies to develop novel diagnostics and therapeutics against fibrosis and cancer.
Collapse
Affiliation(s)
- Saleh Yazdani
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; ScarTec Therapeutics BV, Enschede, The Netherlands.
| |
Collapse
|
11
|
Zeng R, Li B, Huang J, Zhong M, Li L, Duan C, Zeng S, Huang J, Liu W, Lu J, Tang Y, Zhou L, Liu Y, Li J, He Z, Wang Q, Dai Y. Lysophosphatidic Acid is a Biomarker for Peritoneal Carcinomatosis of Gastric Cancer and Correlates with Poor Prognosis. Genet Test Mol Biomarkers 2017; 21:641-648. [PMID: 28910191 DOI: 10.1089/gtmb.2017.0060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Ruolan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Junhui Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Meizuo Zhong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Li Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chaojun Duan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingchen Lu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lingming Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yiping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhuang Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengxi He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youyi Dai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Pavlakis K, Messini I, Yiannou P, Gavresea T, Chrysanthakis D, Hilaris G, Panoskaltsis T. Morcellating uterine mesenchymal tumors: The pathologist's view. J Obstet Gynaecol Res 2017; 43:580-586. [DOI: 10.1111/jog.13240] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/21/2016] [Indexed: 01/16/2023]
Affiliation(s)
- Kitty Pavlakis
- Pathology Department; IASO Women's Hospital; Maroussi Greece
- Pathology Department; National and Kapodistrian University of Athens; Athens, Greece
| | - Irini Messini
- Pathology Department; IASO Women's Hospital; Maroussi Greece
| | - Petros Yiannou
- Pathology Department; IASO Women's Hospital; Maroussi Greece
| | | | | | - Georgios Hilaris
- IASO Women's Hospital Athens Greece and Stanford University Hospital; Stanford California USA
- Stanford University Hospital; Stanford California USA
| | - Theodoros Panoskaltsis
- 2nd Department of Obstetrics and Gynecology; Medical School National and Kapodistrian University of Athens; Athens, Greece
| |
Collapse
|
13
|
Okazaki M, Fushida S, Harada S, Tsukada T, Kinoshita J, Oyama K, Miyashita T, Ninomiya I, Ohta T. Establishing a xenograft mouse model of peritoneal dissemination of gastric cancer with organ invasion and fibrosis. BMC Cancer 2017; 17:23. [PMID: 28056854 PMCID: PMC5217597 DOI: 10.1186/s12885-016-2991-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 12/13/2016] [Indexed: 01/26/2023] Open
Abstract
Background The clinical prognosis of gastric cancer with peritoneal dissemination is poor because of its chemoresistance and rich fibrosis. While several gastric cancer cell lines have been used to establish models of peritoneal dissemination by intraperitoneal injection, most peritoneal tumors that form adopt a medullary pattern in microscopic appearance. This histological finding for the model differs from that in the clinical situation. This study was performed to demonstrate the contribution of human peritoneal mesothelial cells (HPMCs) to fibrotic tumor formation and to establish a new xenograft model with high potential for peritoneal dissemination with organ invasion and extensive fibrosis. Methods We established four types of xenograft model: i) intraperitoneal injection of MKN45-P cells alone (control group), ii) injection of MKN45-P cells co-cultured with HPMCs (co-cultured group), iii) scratching the parietal peritoneum (parietal group), and iv) scratching the visceral peritoneum (visceral group) with a cotton swab before injection of co-cultured cells. Fibrosis, α-smooth muscle actin expression, and organ invasion by tumor cells were all assessed by immunohistochemical examination. Results All mice developed abdominal swelling with peritoneal tumors and bloody ascites. Tumors of the control and co-cultured groups were not invasive or fibrotic. Contrastingly, tumors of the scratch groups exhibited rich stromal fibrosis and possessed increased α-smooth muscle actin (α-SMA) expression. In particular, the visceral group showed edematous and spreading tumors invading the intestinal wall. Conclusion We established a model of peritoneal dissemination with organ invasion and stromal fibrosis. Formation of peritoneal dissemination required a favorable environment for cell adhesion, invasion, and growth. This model may be useful for analyzing the pathogenesis and treatment of peritoneal dissemination of gastric cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2991-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mitsuyoshi Okazaki
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan.
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Tomoya Tsukada
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Katsunobu Oyama
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8641, Ishikawa, Japan
| |
Collapse
|
14
|
Tanaka M, Kuriyama S, Itoh G, Maeda D, Goto A, Tamiya Y, Yanagihara K, Yashiro M, Aiba N. Mesothelial Cells Create a Novel Tissue Niche That Facilitates Gastric Cancer Invasion. Cancer Res 2016; 77:684-695. [PMID: 27895076 DOI: 10.1158/0008-5472.can-16-0964] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
Peritoneal mesothelial cells (PMC) cover organ surfaces in the abdominal cavity. In this study, lineage tracing revealed that the PMCs guide cancer cell invasion in the gastric wall and in peritoneal metastatic lesions. Serosal PMCs covering the stomach surface entered the gastric wall to create a novel niche that favored gastric cancer cell invasion. PMC infiltration was induced by incorporation of cancer cell-derived, Wnt3a-containing extracellular vesicles. Infiltrated PMCs in turn promoted subserosal invasion of cancer cells. Mutual attraction between cancer cells and PMCs accelerated tumor invasion in the gastric wall, and PMC-led cancer cell invasion in disseminated tumors within the abdominal wall and diaphragm. Addition of the carboxyl terminus of Dickkopf-1 attenuated directional invasion of PMCs toward cancer cells both in vitro and in the gastric wall in vivo PMCs were sensitive to the aldehyde dehydrogenase (ALDH) inhibitor disulfiram (DSF), as ALDH activity is elevated in PMCs. Wnt3a upregulated ALDH, and addition of DSF inhibited the invasive properties of PMCs, whereas DSF pretreatment suppressed gastric infiltration of PMCs and subserosal invasion by cancer cells. Our results suggest that stabilization of PMCs may become an effective therapy for the prevention of local invasion and metastasis of gastric cancer. Cancer Res; 77(3); 684-95. ©2016 AACR.
Collapse
Affiliation(s)
- Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Daichi Maeda
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yutaro Tamiya
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, Akita, Japan
| | - Kazuyoshi Yanagihara
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | - Namiko Aiba
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
15
|
Okazaki M, Fushida S, Harada S, Tsukada T, Kinoshita J, Oyama K, Tajima H, Ninomiya I, Fujimura T, Ohta T. The Angiotensin II type 1 receptor blocker candesartan suppresses proliferation and fibrosis in gastric cancer. Cancer Lett 2014; 355:46-53. [DOI: 10.1016/j.canlet.2014.09.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 08/28/2014] [Accepted: 09/10/2014] [Indexed: 12/14/2022]
|
16
|
Shinbo T, Fushida S, Tsukada T, Harada S, Kinoshita J, Oyama K, Okamoto K, Ninomiya I, Takamura H, Kitagawa H, Fujimura T, Yashiro M, Hirakawa K, Ohta T. Protein-bound polysaccharide K suppresses tumor fibrosis in gastric cancer by inhibiting the TGF-β signaling pathway. Oncol Rep 2014; 33:553-8. [PMID: 25435013 PMCID: PMC4306268 DOI: 10.3892/or.2014.3636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/07/2014] [Indexed: 12/22/2022] Open
Abstract
Peritoneal carcinomatosis (PC) is the most frequent metastatic pattern of gastric cancer and its prognosis is extremely poor. PC is characterized by rich fibrosis and the development of obstructive disorders such as ileus, jaundice and hydronephrosis. Epithelial-mesenchymal transition (EMT) is one of the major causes of tissue fibrosis and transforming growth factor β (TGF-β) has a pivotal function in the progression of EMT. Protein-bound polysaccharide K (PSK) is a biological response modifier that can modulate the TGF-β/Smad signaling pathway in vitro. In the present study, we established a fibrotic tumor model using human peritoneal mesothelial cells (HPMCs) and a human gastric cancer cell line to evaluate whether PSK attenuates tumor fibrosis. HPMCs exposed to PSK did not undergo the morphological change from a cobblestone-like pattern to a spindle-shape pattern normally induced by treatment with TGF-β. Immunofluorescence further demonstrated that PSK suppressed TGF-β-induced overexpression of α-SMA in the HPMCs. We further showed that HPMCs contributed to the proliferation of tumor fibrosis by using a mouse xenograft model. Additionally, PSK treatment of these mice significantly reduced the area of observable tumor fibrosis. These results suggest that seeded cancer cells transformed HPMCs into myofibroblast-like cells through their release of TGF-β in the microenvironment, facilitating the development of fibrous tumors in organs covered with HPMCs. Therefore, our study indicates that PSK has potential utility as an anti-fibrotic agent in the treatment of gastric cancer patients with PC.
Collapse
Affiliation(s)
- Toshifumi Shinbo
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tomoya Tsukada
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Katsunobu Oyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Koichi Okamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hirohisa Kitagawa
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Takeshi Fujimura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University, Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Kousei Hirakawa
- Department of Surgical Oncology, Osaka City University, Graduate School of Medicine, Abeno-ku, Osaka 545-8585, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| |
Collapse
|
17
|
Satoyoshi R, Aiba N, Yanagihara K, Yashiro M, Tanaka M. Tks5 activation in mesothelial cells creates invasion front of peritoneal carcinomatosis. Oncogene 2014; 34:3176-87. [PMID: 25088196 DOI: 10.1038/onc.2014.246] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 06/06/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022]
Abstract
Scirrhous gastric cancer is frequently associated with peritoneal dissemination, and the interaction of cancer cells with peritoneal mesothelial cells (PMCs) is crucial for the establishment of the metastasis in the peritoneum. Although cells derived from PMCs are detected within tumors of peritoneal carcinomatosis, how PMCs are incorporated into tumor architecture is not understood. The present study shows that PMCs create the invasion front of peritoneal carcinomatosis, which depends on activation of Tks5 in PMCs. In peritoneal tumor implants, PMCs represent majority of cells located at the invasive edge of the cancer tissue. Exogenously implanted PMCs and host PMCs aggressively invade into abdominal wall upon the peritoneal inoculation of cancer cells, and PMCs locate ahead of cancer cells in the direction of invasion. Tks5, a substrate of Src kinase, is predominantly expressed in the PMCs of cancer tissue, and promotes the invasion of PMCs and cancer cells. Expression and activation of Tks5 was induced in PMCs following their exposure to gastric cancer cells, and increased Tks5 expression was detected in PMCs located at the invasion front. Reduced Tks5 expression in PMCs blocked PMC invasion, which in turn prevents cancer cell invasion both in vitro and in vivo. The peritoneal dissemination of gastric cancer was significantly increased by mixing cancer cells and PMCs, and was suppressed by knockdown of Tks5 in PMCs. These results suggest that cancer-activated PMCs create invasion front by guiding cancer cells. Signaling leading to Tks5 activation in PMCs may be a suitable therapeutic target for prevention of peritoneal carcinomatosis.
Collapse
Affiliation(s)
- R Satoyoshi
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - N Aiba
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - K Yanagihara
- Division of Translational Research, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - M Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - M Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
18
|
Expression of the miR200 Family of microRNAs in Mesothelial Cells Suppresses the Dissemination of Ovarian Cancer Cells. Mol Cancer Ther 2014; 13:2081-91. [DOI: 10.1158/1535-7163.mct-14-0135] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Tumor-associated mesothelial cells are negative prognostic factors in gastric cancer and promote peritoneal dissemination of adherent gastric cancer cells by chemotaxis. Tumour Biol 2014; 35:6105-11. [PMID: 24615523 DOI: 10.1007/s13277-014-1808-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022] Open
Abstract
Peritoneal dissemination is highly frequent in gastric cancer. Damage to human peritoneal mesothelial cell (HPMC) barriers provokes gastric cancer peritoneal dissemination (GCPD), the key events during GCPD, is characterized by fibroblastic development. In this study, we have studied the association between fibroblast activation protein (FAP) expression in peritoneum and the pathological features of the primary tumor. The clinical prognosis of gastric cancer patients was evaluated according to FAP expression. In a gastric cancer cell-HPMC co-culture system, expression of E-cadherin, α-smooth muscle actin, and FAP were evaluated by Western blotting. Gastric cancer cell migration and adhesion to HPMC were also assayed. Our results showed positive peritoneal staining of FAP in 36/86 cases (41.9 %), which was associated with a higher TNM stage in primary gastric cancer and higher incidence of GCPD (both p<0.05). Survival analysis showed FAP expression was an independent prognostic factor of poor survival (p=0.02). Peritoneum of FAP-positive expression exhibited a distinct fibrotic development and expressed higher level of the mesenchymal marker α-SMA, which was confirmed by the in vitro Western blot assay. In HPMC and gastric cancer cell adherence assay, SGC-7901 cells preferentially adhered to TA-HPMC at different cell densities (both p<0.05). Additionally, SGC-7901 cells were more prone to chemotaxis by FAP-expressed tumor-associated-human peritoneal mesothelial cells (TA-HPMC) compared with HPMC co-cultured with normal gastric glandular epithelial cells in a time-dependent manner (both p<0.05). Our study indicated a positive correlation between peritoneum FAP expression and GCPD. FAP-expressed TA-HPMC might be an important cellular component and instigator of GCPD.
Collapse
|
20
|
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: 1.8] [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
|
21
|
Miao ZF, Zhao TT, Wang ZN, Miao F, Xu YY, Mao XY, Gao J, Wu JH, Liu XY, You Y, Xu H, Xu HM. Transforming growth factor-beta1 signaling blockade attenuates gastric cancer cell-induced peritoneal mesothelial cell fibrosis and alleviates peritoneal dissemination both in vitro and in vivo. Tumour Biol 2013; 35:3575-83. [PMID: 24347485 DOI: 10.1007/s13277-013-1472-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022] Open
Abstract
Peritoneal dissemination is the most frequent metastatic pattern of advanced gastric cancer and the main cause of death in gastric cancer patients. Transforming growth factor-beta1 (TGF- ß1), one of the most potent fibrotic stimuli for human peritoneal mesothelial cells, has been shown to play an important role in this process. In this study, we investigated the effect of TGF- ß1 signaling blockade in gastric cancer cell (GCC)-induced human peritoneal mesothelial cell (HPMC) fibrosis. HPMCs were cocultured with the high TGF- ß1 expressing GCC line SGC-7901 and various TGF- ß1 signaling inhibitors or SGC-7901 transfected with TGF-ß1-specific siRNA. HPMC fibrosis was monitored on the basis of morphology. Expression of the epithelial cell marker, E-cadherin, and the mesenchymal marker, α-smooth muscle actin (α-SMA), was evaluated by Western blotting and immunofluorescence confocal imaging. GCC adhesion to HPMC was also assayed. In nude mouse tumor model, the peritoneal fibrotic status was monitored by immunofluorescent confocal imaging and Masson's trichrome staining; formation of metastatic nodular and ascites fluid was also evaluated. Our study demonstrated that GCC expressing high levels of TGF-ß1 induced HMPC fibrosis, which is characterized by both upregulation of E-cadherin and downregulation of α-SMA. Furthermore, HPMC monolayers fibrosis was reversed by TGF- ß1 signaling blockade. In vivo, the TGF- ß1 receptor inhibitor SB-431542 partially attenuated early-stage gastric cancer peritoneal dissemination (GCPD). In conclusion, our study confirms the significance of TGFß1 signaling blockade in attenuating GCPD and may provide a therapeutic target for clinical therapy.
Collapse
Affiliation(s)
- Zhi-Feng Miao
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, No. 155 North Nanjing Street, Heping District, Shenyang, Liaoning Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|