101
|
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: 52] [Impact Index Per Article: 7.4] [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
|
102
|
Dasari S, Fang Y, Mitra AK. Cancer Associated Fibroblasts: Naughty Neighbors That Drive Ovarian Cancer Progression. Cancers (Basel) 2018; 10:cancers10110406. [PMID: 30380628 PMCID: PMC6265896 DOI: 10.3390/cancers10110406] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 01/02/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, and patient prognosis has not improved significantly over the last several decades. In order to improve therapeutic approaches and patient outcomes, there is a critical need for focused research towards better understanding of the disease. Recent findings have revealed that the tumor microenvironment plays an essential role in promoting cancer progression and metastasis. The tumor microenvironment consists of cancer cells and several different types of normal cells recruited and reprogrammed by the cancer cells to produce factors beneficial to tumor growth and spread. These normal cells present within the tumor, along with the various extracellular matrix proteins and secreted factors, constitute the tumor stroma and can compose 10–60% of the tumor volume. Cancer associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment, and play a critical role in promoting many aspects of tumor function. This review will describe the various hypotheses about the origin of CAFs, their major functions in the tumor microenvironment in ovarian cancer, and will discuss the potential of targeting CAFs as a possible therapeutic approach.
Collapse
Affiliation(s)
- Subramanyam Dasari
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47401, USA.
| | - Yiming Fang
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47401, USA.
| | - Anirban K Mitra
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47401, USA.
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
103
|
Peng Y, Kajiyama H, Yuan H, Nakamura K, Yoshihara M, Yokoi A, Fujikake K, Yasui H, Yoshikawa N, Suzuki S, Senga T, Shibata K, Kikkawa F. PAI-1 secreted from metastatic ovarian cancer cells triggers the tumor-promoting role of the mesothelium in a feedback loop to accelerate peritoneal dissemination. Cancer Lett 2018; 442:181-192. [PMID: 30429105 DOI: 10.1016/j.canlet.2018.10.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 10/01/2018] [Accepted: 10/16/2018] [Indexed: 10/28/2022]
Abstract
The mesothelium, covered by a continuous monolayer of mesothelial cells, is the first protective barrier against metastatic ovarian cancer. However, mesothelial cells release tumor-promoting factors that accelerate the process of peritoneal metastasis. We identified cancer-associated mesothelial cells (CAMs) that had tumor-promoting potential. Here, we found that plasminogen activator inhibitor-1 (PAI-1) induced the formation of CAMs, after which CAMs increasingly secreted the oncogenic factors interleukin-8 (IL-8) and C-X-C motif chemokine ligand 5 (CXCL5), further promoting the metastasis of ovarian cancer cells in a feedback loop. After the formation of CAMs, PAI-1 activated the nuclear factor kappa B (NFκB) pathway in the CAMs, thus transcriptionally upregulating the expression of the downstream NFκB targets IL-8 and CXCL5. Moreover, PAI-1 correlated with peritoneal metastasis in ovarian cancer patients and indicated a poor prognosis. In both ex vivo and in vivo models, after PAI-1 expression was knocked down, the metastasis of ovarian cancer cells decreased significantly. Therefore, targeting PAI-1 may provide a potential target for future therapeutics to prevent the formation of CAMs and alleviate peritoneal metastasis in ovarian cancer patients.
Collapse
Affiliation(s)
- Yang Peng
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan.
| | - Hong Yuan
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Kae Nakamura
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Kayo Fujikake
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Hiroaki Yasui
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Shiro Suzuki
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| | - Takeshi Senga
- Department of Internal Medicine, Yahagigawa Hospital, Anjyo, 444-1164, Aichi, Japan
| | - Kiyosumi Shibata
- Department of Obstetrics and Gynecology, Banbuntane Hotokukai Hospital, Fujita Health University, Nakagawa-ku, Nagoya, 454-8509, Aichi, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
104
|
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: 54] [Impact Index Per Article: 7.7] [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
|
105
|
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: 154] [Impact Index Per Article: 22.0] [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
|
106
|
Hilliard TS. The Impact of Mesothelin in the Ovarian Cancer Tumor Microenvironment. Cancers (Basel) 2018; 10:E277. [PMID: 30134520 PMCID: PMC6162689 DOI: 10.3390/cancers10090277] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/17/2018] [Accepted: 08/18/2018] [Indexed: 01/14/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological disease among U.S. women. Poor 5-year survival rates (<30%) are due to presentation of most women at diagnosis with advanced stage disease with widely disseminated intraperitoneal metastasis. However, when diagnosed before metastatic propagation the overall 5-year survival rate is >90%. Metastasizing tumor cells grow rapidly and aggressively attach to the mesothelium of all organs within the peritoneal cavity, including the parietal peritoneum and the omentum, producing secondary lesions. In this review, the involvement of mesothelin (MSLN) in the tumor microenvironment is discussed. MSLN, a 40kDa glycoprotein that is overexpressed in many cancers including ovarian and mesotheliomas is suggested to play a role in cell survival, proliferation, tumor progression, and adherence. However, the biological function of MSLN is not fully understood as MSLN knockout mice do not present with an abnormal phenotype. Conversely, MSLN has been shown to bind to the ovarian cancer antigen, CA-125, and thought to play a role in the peritoneal diffusion of ovarian tumor cells. Although the cancer-specific expression of MSLN makes it a potential therapeutic target, more studies are needed to validate the role of MSLN in tumor metastasis.
Collapse
Affiliation(s)
- Tyvette S Hilliard
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
| |
Collapse
|
107
|
Ghoneum A, Afify H, Salih Z, Kelly M, Said N. Role of tumor microenvironment in the pathobiology of ovarian cancer: Insights and therapeutic opportunities. Cancer Med 2018; 7:5047-5056. [PMID: 30133163 PMCID: PMC6198242 DOI: 10.1002/cam4.1741] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/15/2018] [Accepted: 07/30/2018] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is the fifth most common cancer affecting women and at present, stands as the most lethal gynecologic malignancy. The poor disease outcome is due to the nonspecific symptoms and the lack of effective treatment at advanced stages. Thus, it is of utmost importance to understand ovarian carcinoma through several lenses and to dissect the role that the unique peritoneal tumor microenvironment plays in ovarian cancer progression and metastasis. This review seeks to highlight several determinants of this unique tumor microenvironment, their influence on disease outcome and ongoing clinical trials targeting these determinants.
Collapse
Affiliation(s)
- Alia Ghoneum
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Hesham Afify
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Ziyan Salih
- Department of Pathology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Michael Kelly
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Neveen Said
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, North Carolina.,Department of Pathology, Wake Forest University School of Medicine, Winston Salem, North Carolina.,Department of Urology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
108
|
McKenzie AJ, Hicks SR, Svec KV, Naughton H, Edmunds ZL, Howe AK. The mechanical microenvironment regulates ovarian cancer cell morphology, migration, and spheroid disaggregation. Sci Rep 2018; 8:7228. [PMID: 29740072 PMCID: PMC5940803 DOI: 10.1038/s41598-018-25589-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/24/2018] [Indexed: 01/13/2023] Open
Abstract
There is growing appreciation of the importance of the mechanical properties of the tumor microenvironment on disease progression. However, the role of extracellular matrix (ECM) stiffness and cellular mechanotransduction in epithelial ovarian cancer (EOC) is largely unknown. Here, we investigated the effect of substrate rigidity on various aspects of SKOV3 human EOC cell morphology and migration. Young’s modulus values of normal mouse peritoneum, a principal target tissue for EOC metastasis, were determined by atomic force microscopy (AFM) and hydrogels were fabricated to mimic these values. We find that cell spreading, focal adhesion formation, myosin light chain phosphorylation, and cellular traction forces all increase on stiffer matrices. Substrate rigidity also positively regulates random cell migration and, importantly, directional increases in matrix tension promote SKOV3 cell durotaxis. Matrix rigidity also promotes nuclear translocation of YAP1, an oncogenic transcription factor associated with aggressive metastatic EOC. Furthermore, disaggregation of multicellular EOC spheroids, a behavior associated with dissemination and metastasis, is enhanced by matrix stiffness through a mechanotransduction pathway involving ROCK, actomyosin contractility, and FAK. Finally, this pattern of mechanosensitivity is maintained in highly metastatic SKOV3ip.1 cells. These results establish that the mechanical properties of the tumor microenvironment may play a role in EOC metastasis.
Collapse
Affiliation(s)
- Andrew J McKenzie
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Stephanie R Hicks
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Kathryn V Svec
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Hannah Naughton
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Zöe L Edmunds
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Alan K Howe
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States.
| |
Collapse
|
109
|
Mikuła-Pietrasik J, Stryczyński Ł, Uruski P, Tykarski A, Książek K. Procancerogenic activity of senescent cells: A case of the peritoneal mesothelium. Ageing Res Rev 2018; 43:1-9. [PMID: 29355719 DOI: 10.1016/j.arr.2018.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/13/2018] [Accepted: 01/16/2018] [Indexed: 01/18/2023]
Abstract
Human peritoneal mesothelial cells belong to a narrow group of somatic cells in which both the triggers and the mechanisms of senescence have already been well defined. Importantly, senescent mesothelial cells have been found in the peritoneal cavity in vivo. From a clinical point of view, peritoneal mesothelial cells have been recognized as playing a critical role in the intraperitoneal development of tumor metastases. The pro-cancerogenic behavior of mesothelial cells is even more pronounced when the cells exhaust their proliferative capacity and become senescent. In this review, we summarize the current state of art regarding the contribution of peritoneal mesothelial cells in the progression of ovarian, colorectal, and pancreatic carcinomas, with particular attention paid to the cancer-promoting activity of their senescent counterparts. Moreover, we delineate the mechanisms, mediators, and signaling pathways that are engaged by the senescent mesothelial cells to support such vital elements of cancer progression as adhesion, proliferation, migration, invasion, epithelial-mesenchymal transition, and angiogenesis. Finally, we discuss the experimental evidence regarding both natural and synthetic compounds that may either prevent or restrict cancer development by delaying senescence of mesothelial cells.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Łukasz Stryczyński
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| |
Collapse
|
110
|
Ghoneum A, Afify H, Salih Z, Kelly M, Said N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget 2018; 9:22832-22849. [PMID: 29854318 PMCID: PMC5978268 DOI: 10.18632/oncotarget.25126] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/21/2018] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is the fifth most common cancer affecting the female population and at present, stands as the most lethal gynecologic malignancy. Poor prognosis and low five-year survival rate are attributed to nonspecific symptoms and below par diagnostic criteria at early phases along with a lack of effective treatment at advanced stages. It is thus of utmost importance to understand ovarian carcinoma through several lenses including its molecular pathogenesis, epidemiology, histological subtypes, hereditary factors, diagnostic approaches and methods of treatment. Above all, it is crucial to dissect the role that the unique peritoneal tumor microenvironment plays in ovarian cancer progression and metastasis. This review seeks to highlight several important aspects of ovarian cancer pathobiology as a means to provide the necessary background to approach ovarian malignancies in the future.
Collapse
Affiliation(s)
- Alia Ghoneum
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Hesham Afify
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Ziyan Salih
- Department of Cancer Pathology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Michael Kelly
- Department of Cancer Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| | - Neveen Said
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Department of Cancer Pathology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
- Department of Cancer Urology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
111
|
Wei M, Yang T, Chen X, Wu Y, Deng X, He W, Yang J, Wang Z. Malignant ascites-derived exosomes promote proliferation and induce carcinoma-associated fibroblasts transition in peritoneal mesothelial cells. Oncotarget 2018; 8:42262-42271. [PMID: 28178689 PMCID: PMC5522065 DOI: 10.18632/oncotarget.15040] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/13/2017] [Indexed: 02/05/2023] Open
Abstract
Malignant ascites-derived exosomes have been demonstrated to participate in tumor metastasis. In peritoneal metastasis, normal mesothelial cells (MCs) can be converted into carcinoma-associated fibroblasts (CAFs) by mesothelial-mesenchymal transition (MMT). Herein, we evaluated the effect of malignant ascites-derived exosomes on peritoneal MCs in vitro and in vivo experiments to determine whether exosomes could educate MCs and contribute to peritoneal metastasis. Under the treatment of ascites-derived exosomes, peritoneal MCs showed increased ability to proliferate and migrate. Expression of CAFs specific proteins markers in MCs, including fibroblast activation protein (FAP), alpha-smooth muscle actin (α-SMA), and fibronectin, were increased after treatment of exosomes. In clinical samples test, TGF-β1 was found to be overexpressed in both malignant ascites and malignant ascites-derived exosomes, and the high volume of TGF-β1 may be responsible for peritoneum fibrosis. In addition, exosomes can increase xenograft tumor growth by suppressing the inhibitive ability on tumor cells by MCs. Besides, CAFs specific proteins markers including FAP, α-SMA, and vimentin were increased in clinical peritoneal biopsies. The immunohistochemical staining for mice tumor biopsies also revealed increased expression of fibronectin and FAP, along with decreased expression of E-cadherin and VCAM-1 after exosomes treatment. Thus, malignant ascites-derived exosomes may be of importance in the development of peritoneal metastasis by facilitating MCs to proliferate and convert into CAFs by TGF-β1 induced MMT.
Collapse
Affiliation(s)
- Mingtian Wei
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tinghan Yang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangzheng Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China.,Department of Liver Surgery and Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yangping Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangbing Deng
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wanbin He
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ziqiang Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
112
|
Enomoto Y, Matsushima S, Meguro S, Kawasaki H, Kosugi I, Fujisawa T, Enomoto N, Inui N, Nakamura Y, Suda T, Iwashita T. Podoplanin-positive myofibroblasts: a pathological hallmark of pleuroparenchymal fibroelastosis. Histopathology 2018; 72:1209-1215. [PMID: 29468722 DOI: 10.1111/his.13494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/15/2018] [Indexed: 01/28/2023]
Abstract
Pathological differential diagnoses of pleuroparenchymal fibroelastosis (PPFE) include usual interstitial pneumonia (UIP) and pulmonary apical cap (PAC); however, there are no specific immunostaining makers to distinguish between these diseases. We performed immunohistochemistry using several pleural mesothelial cell-related markers, including cytokeratin-5/6, CAM5.2, WT-1, calretinin, desmin and podoplanin, for PPFE (n = 4), UIP (n = 10) and PAC (n = 3) lung sections. Among the examined markers, in PPFE and PAC lungs podoplanin commonly showed positivity for spindle cells both in thickened pleura and subpleural fibroelastosis lesions; these cells were also stained with α-smooth muscle actin, a marker of myofibroblasts. However, even in elastic fibre-rich cases, UIP lungs did not show such podoplanin-positive myofibroblasts in pleura/subpleura and fibroblastic foci. These findings were also verified using immunofluorescence. By contrast, immunohistochemically as well as morphologically, the difference between PPFE and PAC was not apparent. The presence of podoplanin-positive myofibroblasts could be a pathological hallmark of PPFE, suggesting a pathogenic process distinct from UIP but common to PAC.
Collapse
Affiliation(s)
- Yasunori Enomoto
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan.,Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Sayomi Matsushima
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan.,Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shiori Meguro
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Hideya Kawasaki
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
113
|
Bozzi F, Brich S, Dagrada GP, Negri T, Conca E, Cortelazzi B, Belfiore A, Perrone F, Gualeni AV, Gloghini A, Cabras A, Brenca M, Maestro R, Zaffaroni N, Casali P, Bertulli R, Deraco M, Pilotti S. Epithelioid peritoneal mesothelioma: a hybrid phenotype within a mesenchymal-epithelial/epithelial-mesenchymal transition framework. Oncotarget 2018; 7:75503-75517. [PMID: 27705913 PMCID: PMC5342756 DOI: 10.18632/oncotarget.12262] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/13/2016] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to reconsider the biological characteristics of epithelioid malignant peritoneal mesothelioma (E-MpM) in the light of new concepts about epithelial mesenchymal transition and mesenchymal epithelial reverse transition (EMT/MErT) and the role of epigenetic reprogramming in this context. To this end we profiled surgical specimens and derived cells cultures by a number of complementary approaches i.e. immunohistochemistry, immunofluorescence, in situ hybridization, biochemistry, pluripotent stem cell arrays, treatments with cytokines, growth factors and specific inhibitors.The analyses of the surgical specimens showed that i) EZH2 is expressed throughout the spectrum of MpM, ii) that E-MpM (including the high-grade undifferentiated form) are characterised by c-MYC and miRNA 17-5p expression, and iii) that progression to sarcomatoid MpM is dictated by EMT regulators. They also showed that E-MpM expressed c-MET and are enriched in E- and P-cadherins- and VEGFR2-expressing CSCs, thus strongly supporting a role for MErT reprogramming in endowing E-MpM tumour cells with stemness and plasticity, and hence with a drug resistant phenotype. The cell culture-based experiments confirmed the stemness traits and plasticity of E-MpM, and support the view that EZH2 is a druggable target in this tumor.
Collapse
Affiliation(s)
- Fabio Bozzi
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Brich
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,MOSE-DEA University of Trieste, Trieste, Italy
| | - Gian Paolo Dagrada
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tiziana Negri
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Conca
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Cortelazzi
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonino Belfiore
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ambra Vittoria Gualeni
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonello Cabras
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Brenca
- Experimental Oncology 1, Centro di Riferimento Oncologico, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Roberta Maestro
- Experimental Oncology 1, Centro di Riferimento Oncologico, CRO Aviano National Cancer Institute, Aviano, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Casali
- Adult Mesenchymal Tumor Medical Oncology Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rossella Bertulli
- Adult Mesenchymal Tumor Medical Oncology Unit, Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marcello Deraco
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
114
|
Mikuła-Pietrasik J, Uruski P, Tykarski A, Książek K. The peritoneal "soil" for a cancerous "seed": a comprehensive review of the pathogenesis of intraperitoneal cancer metastases. Cell Mol Life Sci 2018; 75:509-525. [PMID: 28956065 PMCID: PMC5765197 DOI: 10.1007/s00018-017-2663-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/29/2017] [Accepted: 09/20/2017] [Indexed: 01/02/2023]
Abstract
Various types of tumors, particularly those originating from the ovary and gastrointestinal tract, display a strong predilection for the peritoneal cavity as the site of metastasis. The intraperitoneal spread of a malignancy is orchestrated by a reciprocal interplay between invading cancer cells and resident normal peritoneal cells. In this review, we address the current state-of-art regarding colonization of the peritoneal cavity by ovarian, colorectal, pancreatic, and gastric tumors. Particular attention is paid to the pro-tumoral role of various kinds of peritoneal cells, including mesothelial cells, fibroblasts, adipocytes, macrophages, the vascular endothelium, and hospicells. Anatomo-histological considerations on the pro-metastatic environment of the peritoneal cavity are presented in the broader context of organ-specific development of distal metastases in accordance with Paget's "seed and soil" theory of tumorigenesis. The activity of normal peritoneal cells during pivotal elements of cancer progression, i.e., adhesion, migration, invasion, proliferation, EMT, and angiogenesis, is discussed from the perspective of well-defined general knowledge on a hospitable tumor microenvironment created by the cellular elements of reactive stroma, such as cancer-associated fibroblasts and macrophages. Finally, the paper addresses the unique features of the peritoneal cavity that predispose this body compartment to be a niche for cancer metastases, presents issues that are topics of an ongoing debate, and points to areas that still require further in-depth investigations.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland.
| |
Collapse
|
115
|
Asano Y, Odagiri T, Oikiri H, Matsusaki M, Akashi M, Shimoda H. Construction of artificial human peritoneal tissue by cell-accumulation technique and its application for visualizing morphological dynamics of cancer peritoneal metastasis. Biochem Biophys Res Commun 2017; 494:213-219. [DOI: 10.1016/j.bbrc.2017.10.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023]
|
116
|
van Baal JOAM, van Noorden CJF, Nieuwland R, Van de Vijver KK, Sturk A, van Driel WJ, Kenter GG, Lok CAR. Development of Peritoneal Carcinomatosis in Epithelial Ovarian Cancer: A Review. J Histochem Cytochem 2017; 66:67-83. [PMID: 29164988 DOI: 10.1369/0022155417742897] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Epithelial ovarian cancer (EOC) metastasizes intra-abdominally with often numerous, superficial, small-sized lesions. This so-called peritoneal carcinomatosis is difficult to treat, and peritoneal recurrences are frequently observed, leading to a poor prognosis. Underlying mechanisms of interactions between EOC and peritoneal cells are incompletely understood. This review summarizes and discusses the development of peritoneal carcinomatosis from a cell-biological perspective, focusing on characteristics of EOC and peritoneal cells. We aim to provide insight into how peritoneum facilitates tumor adhesion but limits size of lesions and depth of invasion. The development of peritoneal carcinomatosis is a multistep process that requires adaptations of EOC and peritoneal cells. Mechanisms that enable tumor adhesion and growth involve cadherin restructuring on EOC cells, integrin-mediated adhesion, and mesothelial evasion by mechanical forces driven by integrin-ligand interactions. Clinical trials targeting these mechanisms, however, showed only limited effects. Other factors that inhibit tumor growth and deep invasion are virtually unknown. Future studies are needed to elucidate the exact mechanisms that underlie the development and limited growth of peritoneal carcinomatosis. This review on development of peritoneal carcinomatosis of EOC summarizes the current knowledge and its limitations. Clarification of the stepwise process may inspire future research to investigate new treatment approaches of peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Juliette O A M van Baal
- Department of Gynecologic Oncology, Center for Gynecologic Oncology, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Cornelis J F van Noorden
- Cancer Center Amsterdam, Department of Medical Biology, Academic Medical Center, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Koen K Van de Vijver
- Division of Diagnostic Oncology & Molecular Pathology, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Auguste Sturk
- Department of Clinical Chemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Willemien J van Driel
- Department of Gynecologic Oncology, Center for Gynecologic Oncology, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Gemma G Kenter
- Department of Gynecologic Oncology, Center for Gynecologic Oncology, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Christianne A R Lok
- Department of Gynecologic Oncology, Center for Gynecologic Oncology, Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
117
|
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: 127] [Impact Index Per Article: 15.9] [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
|
118
|
Deng G, Qu J, Zhang Y, Che X, Cheng Y, Fan Y, Zhang S, Na D, Liu Y, Qu X. Gastric cancer-derived exosomes promote peritoneal metastasis by destroying the mesothelial barrier. FEBS Lett 2017. [PMID: 28643334 DOI: 10.1002/1873-3468.12722] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
An intact mesothelium serves as a protective barrier to inhibit peritoneal carcinomatosis. Cancer-derived exosomes can mediate directional tumor metastasis; however, little is known about whether gastric cancer-derived exosomes will destroy the mesothelial barrier and promote peritoneal dissemination. Here, we demonstrate that gastric cancer-derived exosomes facilitate peritoneal metastasis by causing mesothelial barrier disruption and peritoneal fibrosis. Injury of peritoneal mesothelial cells elicited by gastric cancer-derived exosomes is through concurrent apoptosis and mesothelial-to-mesenchymal transition (MMT). Additionally, upregulation of p-ERK in peritoneal mesothelial cells is primarily responsible for the MMT while contributing little to apoptosis. Together, these data support the concept that exosomes play a crucial role in remodeling the premetastatic microenvironment and identify a novel mechanism for peritoneal metastasis of gastric carcinoma.
Collapse
Affiliation(s)
- Guang Deng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinglei Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ye Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yu Cheng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Simeng Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Di Na
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
119
|
Jouvin I, Najah H, Pimpie C, Canet Jourdan C, Kaci R, Mirshahi M, Eveno C, Pocard M. Reduction of carcinomatosis risk using icodextrin as a carrier solution of intraperitoneal oxaliplatin chemotherapy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2017; 43:1088-1094. [PMID: 28089175 DOI: 10.1016/j.ejso.2016.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/29/2016] [Accepted: 12/08/2016] [Indexed: 01/14/2023]
Abstract
There is no standard treatment in patients with high risk metachronous peritoneal carcinomatosis (PC) in colonic cancer, as perforated tumour or synchronous ovarian metastasis. Icodextrin 4% (ICDX), presently used to prevent postoperative abdominal adhesions, could inhibit the coactivation of the tumour cells and the microenvironment cells, associated with the development of PC. The aim of this study was to inhibit the formation of the PC in a murine model mimicking surgical situation using ICDX and intraperitoneal (IP) prophylactic chemotherapy. We created a model of growing PC in mice using cells of murine colonic cancer CT26. Cells and treatments were injected simultaneously. Five groups were created: CT26 (control group), CT26 + ICDX (ICDX group), CT26 + chemotherapy (oxaliplatin and 5FU) (chemo group), CT26 + chemotherapy + ICDX (ICDX chemo group), ICDX (toxicity group). At day 15, PC was evaluated with rodents PCI. In the chemo group, PCI was significantly lower than in the control group (3.2 versus 8.4, p = 0.02). ICDX had a synergetic effect on PC with chemotherapy; indeed PCI in ICDX chemo group was lower than in chemo group (1.4 versus 3.2, p = 0.04). There was no morbidity linked to ICDX in toxicity group. Safety of ICDX needs to be verified, particularly on colonic anastomosis before ICDX associated to IP chemotherapy could be used as a preventive treatment of PC in high risk patients. This prophylactic treatment is easy to use and would be administrated at the end of a curative surgery for a colonic cancer.
Collapse
Affiliation(s)
- I Jouvin
- Department of Oncologic & Digestive Surgery, Hospital Lariboisière - AP-HP, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - H Najah
- Department of Oncologic & Digestive Surgery, Hospital Lariboisière - AP-HP, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - C Pimpie
- Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - C Canet Jourdan
- Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - R Kaci
- Department of Anatomopathology, Hôpital Lariboisière - AP-HP, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France
| | - M Mirshahi
- Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - C Eveno
- Department of Oncologic & Digestive Surgery, Hospital Lariboisière - AP-HP, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France
| | - M Pocard
- Department of Oncologic & Digestive Surgery, Hospital Lariboisière - AP-HP, 2 rue Ambroise Paré, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, CART, Carcinomatosis Angiogenesis Translational Research, INSERM U965, F-74575 Paris, France.
| |
Collapse
|
120
|
Rynne-Vidal A, Au-Yeung CL, Jiménez-Heffernan JA, Pérez-Lozano ML, Cremades-Jimeno L, Bárcena C, Cristóbal-García I, Fernández-Chacón C, Yeung TL, Mok SC, Sandoval P, López-Cabrera M. Mesothelial-to-mesenchymal transition as a possible therapeutic target in peritoneal metastasis of ovarian cancer. J Pathol 2017; 242:140-151. [PMID: 28247413 PMCID: PMC5468005 DOI: 10.1002/path.4889] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/28/2016] [Accepted: 02/17/2017] [Indexed: 12/12/2022]
Abstract
Peritoneal dissemination is the primary metastatic route of ovarian cancer (OvCa), and is often accompanied by the accumulation of ascitic fluid. The peritoneal cavity is lined by mesothelial cells (MCs), which can be converted into carcinoma‐associated fibroblasts (CAFs) through mesothelial‐to‐mesenchymal transition (MMT). Here, we demonstrate that MCs isolated from ascitic fluid (AFMCs) of OvCa patients with peritoneal implants also undergo MMT and promote subcutaneous tumour growth in mice. RNA sequencing of AFMCs revealed that MMT‐related pathways – including transforming growth factor (TGF)‐β signalling – are differentially regulated, and a gene signature was verified in peritoneal implants from OvCa patients. In a mouse model, pre‐induction of MMT resulted in increased peritoneal tumour growth, whereas interfering with the TGF‐β receptor reduced metastasis. MC‐derived CAFs showed activation of Smad‐dependent TGF‐β signalling, which was disrupted in OvCa cells, despite their elevated TGF‐β production. Accordingly, targeting Smad‐dependent signalling in the peritoneal pre‐metastatic niche in mice reduced tumour colonization, suggesting that Smad‐dependent MMT could be crucial in peritoneal carcinomatosis. Together, these results indicate that bidirectional communication between OvCa cells and MC‐derived CAFs, via TGF‐β‐mediated MMT, seems to be crucial to form a suitable metastatic niche. We suggest MMT as a possible target for therapeutic intervention and a potential source of biomarkers for improving OvCa diagnosis and/or prognosis. © 2017 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)
- Angela Rynne-Vidal
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain.,Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chi Lam Au-Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - José A Jiménez-Heffernan
- Departamento de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - María Luisa Pérez-Lozano
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Lucía Cremades-Jimeno
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Carmen Bárcena
- Departamento de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Tsz Lun Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pilar Sandoval
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Manuel López-Cabrera
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| |
Collapse
|
121
|
Mikuła-Pietrasik J, Uruski P, Kucińska M, Tykarski A, Książek K. The protective activity of mesothelial cells against peritoneal growth of gastrointestinal tumors: The role of soluble ICAM-1. Int J Biochem Cell Biol 2017; 86:26-31. [PMID: 28323210 DOI: 10.1016/j.biocel.2017.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/12/2017] [Accepted: 03/13/2017] [Indexed: 11/28/2022]
Abstract
In this project we examined how the presence of human peritoneal mesothelial cells (HPMCs) modifies (supports or inhibits) colorectal and pancreatic cancer cell progression in mice peritoneal cavity. Experiments were performed using primary, omentum-derived HPMCs, commercially available colorectal (SW-480) and pancreatic (PSN-1) cancer cells, and immunocompromised SCID mice. Tumor growth within the peritoneal cavity was monitored using bioluminescence. Adhesion of the cancer cells to HPMCs was examined using a fluorescence-based method, while the incidence of apoptosis was quantified using flow cytometry. Experiments showed that SW480 and PSN-1 cells formed tumors in vivo at higher efficiency when they were injected alone than in the presence of HPMCs. In vitro investigations confirmed that firm adhesion of SW480 and PSN-1 cells to HPMCs is mediated by interactions between ICAM-1 and CD43. They also revealed that IL-6 and TNFα up-regulate the expression of cell-bound ICAM-1 and the secretion of soluble ICAM-1 (sICAM-1). The basal release of sICAM-1 by HPMCs positively correlated with the expression of the cell-bound molecule. sICAM-1 inhibited dose-dependently the adhesion of SW480 and PSN-1 cells to HPMCs. Cancer cells that did not adhere to HPMCs displayed increased activity of caspase-3 and -9, increased incidence of apoptosis, and an inability to re-adhesion, as compared with their intact counterparts not exposed to sICAM-1. Our findings indicate that under certain conditions HPMCs are capable of inhibiting growth of gastrointestinal tumors in a mechanism involving the anti-adhesive capabilities of sICAM-1.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Małgorzata Kucińska
- Department of Toxicology, Poznań University of Medical Sciences, Dojazd 30 Str., 60-631 Poznań, Poland.
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848 Poznań, Poland.
| |
Collapse
|
122
|
van Baal J, Van de Vijver K, Nieuwland R, van Noorden C, van Driel W, Sturk A, Kenter G, Rikkert L, Lok C. The histophysiology and pathophysiology of the peritoneum. Tissue Cell 2017; 49:95-105. [DOI: 10.1016/j.tice.2016.11.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 12/14/2022]
|
123
|
Cancer-associated peritoneal mesothelial cells lead the formation of pancreatic cancer peritoneal dissemination. Int J Oncol 2016; 50:457-467. [PMID: 28035373 DOI: 10.3892/ijo.2016.3829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/21/2016] [Indexed: 11/05/2022] Open
Abstract
The interaction between the cancer cells and the peritoneal mesothelial cells (PMCs) plays an important role in the peritoneal dissemination in several types of cancer. However, the role of PMCs in the peritoneal dissemination of pancreatic cancer remains unclear. In the present study, we investigated the interaction between the pancreatic cancer cells (PCCs) and the PMCs in the formation of peritoneal dissemination in vitro and in vivo. The tumor-stromal interaction of PCCs and PMCs significantly enhanced their mobility and invasiveness and enhanced the proliferation and anoikis resistance of PCCs. In a 3D organotypic culture model of peritoneal dissemination, co-culture of PCCs and PMCs significantly increased the cells invading into the collagen gel layer compared with mono-culture of PCCs. PMCs pre-invaded into the collagen gel, remodeled collagen fibers, and increased parallel fiber orientation along the direction of cell invasion. In the tissues of peritoneal dissemination of the KPC (LSL-KrasG12D/+; LSL-Trp53R172H/+;Pdx-1-Cre) transgenic mouse, the monolayer of PMCs was preserved in tumor-free areas, whereas PMCs around the invasive front of peritoneal dissemination proliferated and invaded into the muscle layer. In vivo, intraperitoneal injection of PCCs with PMCs significantly promoted peritoneal dissemination compared with PCCs alone. The present data suggest that the cancer-associated PMCs have important promoting roles in the peritoneal dissemination of PCCs. Therapy targeting cancer-associated PMCs may improve the prognosis of patients with pancreatic cancer.
Collapse
|
124
|
Abstract
Mesothelial cells (MCs) cover the surface of visceral organs and the parietal walls of cavities, and they synthesize lubricating fluids to create a slippery surface that facilitates movement between organs without friction. Recent studies have indicated that MCs play active roles in liver development, fibrosis, and regeneration. During liver development, the mesoderm produces MCs that form a single epithelial layer of the mesothelium. MCs exhibit an intermediate phenotype between epithelial cells and mesenchymal cells. Lineage tracing studies have indicated that during liver development, MCs act as mesenchymal progenitor cells that produce hepatic stellate cells, fibroblasts around blood vessels, and smooth muscle cells. Upon liver injury, MCs migrate inward from the liver surface and produce hepatic stellate cells or myofibroblast depending on the etiology, suggesting that MCs are the source of myofibroblasts in capsular fibrosis. Similar to the activation of hepatic stellate cells, transforming growth factor β induces the conversion of MCs into myofibroblasts. Further elucidation of the biological and molecular changes involved in MC activation and fibrogenesis will contribute to the development of novel approaches for the prevention and therapy of liver fibrosis.
Collapse
Affiliation(s)
- Ingrid Lua
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kinji Asahina
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
125
|
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: 3.0] [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
|
126
|
Sandoval P, Jiménez-Heffernan JA, Guerra-Azcona G, Pérez-Lozano ML, Rynne-Vidal Á, Albar-Vizcaíno P, Gil-Vera F, Martín P, Coronado MJ, Barcena C, Dotor J, Majano PL, Peralta AA, López-Cabrera M. Mesothelial-to-mesenchymal transition in the pathogenesis of post-surgical peritoneal adhesions. J Pathol 2016; 239:48-59. [DOI: 10.1002/path.4695] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/30/2015] [Accepted: 01/26/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Pilar Sandoval
- Centro de Biología Molecular-Severo Ochoa; CSIC. Cantoblanco; Madrid Spain
| | - José A Jiménez-Heffernan
- Departamento de Anatomía Patológica; Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP); Madrid Spain
| | | | | | - Ángela Rynne-Vidal
- Centro de Biología Molecular-Severo Ochoa; CSIC. Cantoblanco; Madrid Spain
| | - Patricia Albar-Vizcaíno
- Unidad de Biología Molecular y Servicio de Nefrología, Hospital Universitario La Princesa; Instituto de Investigación Sanitaria Princesa (IP); Madrid Spain
| | | | - Paloma Martín
- Departamento de Anatomía Patológica; Hospital Universitario Puerta de Hierro; Madrid Spain
| | - María José Coronado
- Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Hospital Puerta de Hierro; Madrid Spain
| | - Carmen Barcena
- Departamento de Anatomía Patológica, Hospital Universitario 12 de Octubre; Madrid Spain
| | | | - Pedro Lorenzo Majano
- Unidad de Biología Molecular y Servicio de Nefrología, Hospital Universitario La Princesa; Instituto de Investigación Sanitaria Princesa (IP); Madrid Spain
| | - Abelardo Aguilera Peralta
- Unidad de Biología Molecular y Servicio de Nefrología, Hospital Universitario La Princesa; Instituto de Investigación Sanitaria Princesa (IP); Madrid Spain
| | | |
Collapse
|
127
|
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
|
128
|
The Mesothelial Origin of Carcinoma Associated-Fibroblasts in Peritoneal Metastasis. Cancers (Basel) 2015; 7:1994-2011. [PMID: 26426054 PMCID: PMC4695872 DOI: 10.3390/cancers7040872] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/14/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023] Open
Abstract
Solid tumors are complex and unstructured organs that, in addition to cancer cells, also contain other cell types. Carcinoma-associated fibroblasts (CAFs) represent an important population in the tumor microenviroment and participate in several stages of tumor progression, including cancer cell migration/invasion and metastasis. During peritoneal metastasis, cancer cells detach from the primary tumor, such as ovarian or gastrointestinal, disseminate through the peritoneal fluid and colonize the peritoneum. Tumor cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity, then colonizing the submesothelial compact zone where CAFs accumulate. CAFs may derive from different sources depending on the surrounding metastatic niche. In peritoneal metastasis, a sizeable subpopulation of CAFs originates from MCs through a mesothelial-to-mesenchymal transition (MMT), which promotes adhesion, invasion, vascularization and subsequent tumor growth. The bidirectional communication between cancer cells and MC-derived CAFs via secretion of a wide range of cytokines, growth factors and extracellular matrix components seems to be crucial for the establishment and progression of the metastasis in the peritoneum. This manuscript provides a comprehensive review of novel advances in understanding how peritoneal CAFs provide cancer cells with a supportive microenvironment, as well as the development of future therapeutic approaches by interfering with the MMT in the peritoneum.
Collapse
|
129
|
Nakamura M, Ono YJ, Kanemura M, Tanaka T, Hayashi M, Terai Y, Ohmichi M. Hepatocyte growth factor secreted by ovarian cancer cells stimulates peritoneal implantation via the mesothelial-mesenchymal transition of the peritoneum. Gynecol Oncol 2015; 139:345-54. [PMID: 26335595 DOI: 10.1016/j.ygyno.2015.08.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/13/2015] [Accepted: 08/17/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE A current working model for the metastatic process of ovarian carcinoma suggests that cancer cells are shed from the ovarian tumor into the peritoneal cavity and attach to the layer of mesothelial cells that line the inner surface of the peritoneum, and several studies suggest that hepatocyte growth factor (HGF) plays an important role in the dissemination of ovarian cancer. Our objectives were to evaluate the HGF expression of ovarian cancer using clinical data and assess the effect of HGF secreted from human ovarian cancer cells to human mesothelial cells. METHODS HGF expression was immunohistochemically evaluated in 165 epithelial ovarian cancer patients arranged as tissue microarrays. HGF expression in four ovarian cancer cell lines was evaluated by using semi-quantitative polymerase chain reaction, Western blotting and enzyme-linked immunosorbent assay. The effect of ovarian cancer cell derived HGF to the human mesothelial cells was assessed by using morphologic analysis, Western blotting and cell invasion assay. The effect of HGF on ovarian cancer metastasis was assessed by using in vivo experimental model. RESULTS The clinical data showed a significantly high correlation between the HGF expression and the cancer stage. The in vivo and in vitro experimental models revealed that HGF secreted by ovarian cancer cells induces the mesothelial-to-mesenchymal transition and stimulates the invasion of mesothelial cells. Furthermore, manipulating the HGF activity affected the degree of dissemination and ascite formation. CONCLUSIONS We demonstrated that HGF secreted by ovarian cancer cells plays an important role in cancer peritoneal implantation.
Collapse
Affiliation(s)
- Michihiko Nakamura
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| | - Yoshihiro J Ono
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan.
| | - Masanori Kanemura
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| | - Masami Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College, 2-7 Daigakumachi, Takatsuki, Osaka 5650781, Japan
| |
Collapse
|
130
|
De Vlieghere E, Verset L, Demetter P, Bracke M, De Wever O. Cancer-associated fibroblasts as target and tool in cancer therapeutics and diagnostics. Virchows Arch 2015; 467:367-82. [PMID: 26259962 DOI: 10.1007/s00428-015-1818-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/21/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are drivers of tumour progression and are considered as a target and a tool in cancer diagnostic and therapeutic applications. An increased abundance of CAFs or CAF signatures are recognized as a bad prognostic marker in several cancer types. Tumour-environment biomimetics strongly improve our understanding of the communication between CAFs, cancer cells and other host cells. Several experimental drugs targeting CAFs are in clinical trials for multiple tumour entities; alternatively, CAFs can be exploited as a tool to characterize the functionality of circulating tumour cells or to capture them as a tool to prevent metastasis. The continuous interaction between tissue engineers, biomaterial experts and cancer researchers creates the possibility to biomimic the tumour-environment and provides new opportunities in cancer diagnostics and management.
Collapse
Affiliation(s)
- Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Laurine Verset
- Departments of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Pieter Demetter
- Departments of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium.
| |
Collapse
|
131
|
Ono YJ, Hayashi M, Tanabe A, Hayashi A, Kanemura M, Terai Y, Ohmichi M. Estradiol-mediated hepatocyte growth factor is involved in the implantation of endometriotic cells via the mesothelial-to-mesenchymal transition in the peritoneum. Am J Physiol Endocrinol Metab 2015; 308:E950-9. [PMID: 25852006 DOI: 10.1152/ajpendo.00573.2014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022]
Abstract
The pathogenesis of endometriosis, a chronic painful gynecological disease characterized by the presence of endometrial tissue located outside of the uterus and often adhering to the peritoneum, is known to be estrogen dependent. However, the precise pathophysiology of endometriosis remains elusive. Recent studies indicate that the epithelial-to-mesenchymal transition (EMT) of human endometrial cells is important for the progression of endometriosis, and another previous study has implicated hepatocyte growth factor (HGF) in endometriosis progression. The aim of the present study was to examine the role of estradiol in the regulation of HGF production and progression of peritoneal endometriosis, focusing on the interactions between the peritoneum and endometriotic cells. Consequently, estradiol was found to promote the proliferation, invasion, and migration of immortalized human endometrial epithelial cells (hEECs) via HGF upregulation, and the estradiol-induced direct binding of estrogen receptor-α to the HGF promoter was confirmed on a chromatin immunoprecipitation (ChIP) assay. Estradiol also induced the EMT in hEECs by promoting HGF production. Furthermore, human mesothelial cells underwent the mesothelial-to-mesenchymal transition (MMT) during culture with estradiol-stimulated hEEC conditioned medium. Importantly, estradiol itself did not induce the MMT, and the estradiol-stimulated hEEC-conditioned medium in the presence of HGF antibodies reversed the MMT process. These results, which were obtained using immortalized hEECs, indicate that estradiol-induced HGF production may play a crucial role in the peritoneal implantation of human endometriotic cells by exerting proliferative and invasive effects via the EMT in hEECs and promoting the MMT in mesothelial cells.
Collapse
Affiliation(s)
- Yoshihiro J Ono
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Masami Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Akiko Tanabe
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Atsushi Hayashi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Masanori Kanemura
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Osaka Medical College, Osaka, Japan
| |
Collapse
|
132
|
De Vlieghere E, Gremonprez F, Verset L, Mariën L, Jones CJ, De Craene B, Berx G, Descamps B, Vanhove C, Remon JP, Ceelen W, Demetter P, Bracke M, De Geest BG, De Wever O. Tumor-environment biomimetics delay peritoneal metastasis formation by deceiving and redirecting disseminated cancer cells. Biomaterials 2015; 54:148-57. [PMID: 25907048 DOI: 10.1016/j.biomaterials.2015.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/22/2022]
Abstract
Peritoneal metastasis is life threatening and is the result of an extensive communication between disseminated cancer cells, mesothelial cells and cancer-associated fibroblasts (CAF). CAFs secrete extracellular matrix (ECM) proteins creating a receptive environment for peritoneal implantation. Considering cancer as an ecosystem may provide opportunities to exploit CAFs to create biomimetic traps to deceive and redirect cancer cells. We have designed microparticles (MP) containing a CAF-derived ECM-surface that is intended to compete with natural niches. CAFs were encapsulated in alginate/gelatine beads (500-750 μm in diameter) functionalised with a polyelectrolyte coating (MP[CAF]). The encapsulated CAFs remain viable and metabolically active (≥35 days), when permanently encapsulated. CAF-derived ECM proteins are retained by the non-biodegradable coating. Adhesion experiments mimicking the environment of the peritoneal cavity show the selective capture of floating cancer cells from different tumor origins by MP[CAF] compared to control MP. MP[CAF] are distributed throughout the abdominal cavity without attachment to intestinal organs and without signs of inflammatory reaction. Intraperitoneal delivery of MP[CAF] and sequential removal redirects cancer cell adhesion from the surgical wound to the MP[CAF], delays peritoneal metastasis formation and prolongs animal survival. Our experiments suggest the use of a biomimetic trap based on tumor-environment interactions to delay peritoneal metastasis.
Collapse
Affiliation(s)
- Elly De Vlieghere
- Laboratory of Experimental Cancer Research, Ghent University, Belgium
| | | | - Laurine Verset
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Belgium
| | - Lore Mariën
- Laboratory of Experimental Cancer Research, Ghent University, Belgium
| | | | - Bram De Craene
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Geert Berx
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Benedicte Descamps
- Department of Electronics and Information Systems, IBiTech, MEDISIP, INFINITY, Ghent University, Belgium
| | - Christian Vanhove
- Department of Electronics and Information Systems, IBiTech, MEDISIP, INFINITY, Ghent University, Belgium
| | - Jean-Paul Remon
- Laboratory of Pharmaceutical Technology, Ghent University, Belgium
| | - Wim Ceelen
- Department of Surgery, Ghent University Hospital, Belgium
| | - Pieter Demetter
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Ghent University, Belgium
| | - Bruno G De Geest
- Laboratory of Pharmaceutical Technology, Ghent University, Belgium.
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Ghent University, Belgium.
| |
Collapse
|
133
|
Lung cancer cells induce senescence and apoptosis of pleural mesothelial cells via transforming growth factor-beta1. Tumour Biol 2014; 36:2657-65. [PMID: 25433501 DOI: 10.1007/s13277-014-2888-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023] Open
Abstract
Pleural dissemination is commonly associated with metastatic advanced lung cancer. The injury of pleural mesothelial cells (PMCs) by soluble factors, such as transforming growth factor-beta1 (TGF-β1), is a major driver of lung cancer pleural dissemination (LCPD). In this study, we examine the effects of TGF-β1 on PMC injury and the ability of TGF-β1 inhibition to alleviate this effect both in vitro and in vivo. PMCs were co-cultured with the high TGF-β1-expressing lung cancer cell line A549 and with various TGF-β1 signaling inhibitors. Expression of cleaved-caspase 3, cleaved-caspase 9, p21, and p16 were evaluated by Western blot and immunofluorescent confocal imaging. Apoptosis was measured by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltrazoliumbromide assay and AnnexinV-propidium iodide (PI) staining. PMC senescence was assessed by staining for senescence-associated β-galactosidase (SA-β-Gal). The ability of lung cancer cells (LCCs) to adhere to injured PMCs was investigated using an LCC-PMC adhesion assay. In our mouse model, PMC injury status was monitored by hematoxylin-eosin (H&E) and Masson's trichrome staining. LCCs expressing high levels of TGF-β1 induce apoptosis and senescence of PMCs in a co-culture system. Injured PMCs adhere to LCCs, which may further promote LCPD. Importantly, PMC monolayer injury could be reversed with TGF-β1 inhibitors. This was consistent with our in vivo data showing that the TGF-β1 inhibitor SB-431542 attenuated PMC barrier injury induced by A549 culture medium in our mouse model. Our study highlights the importance of TGF-β1 signaling in LCPD and establishes this signaling pathway as a potential therapeutic target in the disease.
Collapse
|
134
|
Kenny HA, Chiang CY, White EA, Schryver EM, Habis M, Romero IL, Ladanyi A, Penicka CV, George J, Matlin K, Montag A, Wroblewski K, Yamada SD, Mazar AP, Bowtell D, Lengyel E. Mesothelial cells promote early ovarian cancer metastasis through fibronectin secretion. J Clin Invest 2014; 124:4614-28. [PMID: 25202979 DOI: 10.1172/jci74778] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 07/31/2014] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer (OvCa) metastasizes to organs in the abdominal cavity, such as the omentum, which are covered by a single layer of mesothelial cells. Mesothelial cells are generally thought to be "bystanders" to the metastatic process and simply displaced by OvCa cells to access the submesothelial extracellular matrix. Here, using organotypic 3D cultures, we found that primary human mesothelial cells secrete fibronectin in the presence of OvCa cells. Moreover, we evaluated the tumor stroma of 108 human omental metastases and determined that fibronectin was consistently overexpressed in these patients. Blocking fibronectin production in primary mesothelial cells in vitro or in murine models, either genetically (fibronectin 1 floxed mouse model) or via siRNA, decreased adhesion, invasion, proliferation, and metastasis of OvCa cells. Using a coculture model, we determined that OvCa cells secrete TGF-β1, which in turn activates a TGF-β receptor/RAC1/SMAD-dependent signaling pathway in the mesothelial cells that promotes a mesenchymal phenotype and transcriptional upregulation of fibronectin. Additionally, blocking α5 or β1 integrin function with antibodies reduced metastasis in an orthotopic preclinical model of OvCa metastasis. These findings indicate that cancer-associated mesothelial cells promote colonization during the initial steps of OvCa metastasis and suggest that mesothelial cells actively contribute to metastasis.
Collapse
|
135
|
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: 1.9] [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
|