1
|
Cai H, Wang Z, Tang W, Ke X, Zhao E. Recent advances of the mammalian target of rapamycin signaling in mesenchymal stem cells. Front Genet 2022; 13:970699. [PMID: 36110206 PMCID: PMC9468880 DOI: 10.3389/fgene.2022.970699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase involved in a variety of cellular functions, such as cell proliferation, metabolism, autophagy, survival and cytoskeletal organization. Furthermore, mTOR is made up of three multisubunit complexes, mTOR complex 1, mTOR complex 2, and putative mTOR complex 3. In recent years, increasing evidence has suggested that mTOR plays important roles in the differentiation and immune responses of mesenchymal stem cells (MSCs). In addition, mTOR is a vital regulator of pivotal cellular and physiological functions, such as cell metabolism, survival and ageing, where it has emerged as a novel therapeutic target for ageing-related diseases. Therefore, the mTOR signaling may develop a large impact on the treatment of ageing-related diseases with MSCs. In this review, we discuss prospects for future research in this field.
Collapse
Affiliation(s)
- Huarui Cai
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Zhongze Wang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Wenhan Tang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| | - Xiaoxue Ke
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Xiaoxue Ke, ; Erhu Zhao,
| | - Erhu Zhao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Xiaoxue Ke, ; Erhu Zhao,
| |
Collapse
|
2
|
Irons EE, Cortes Gomez E, Andersen VL, Lau JTY. Bacterial colonization and TH17 immunity are shaped by intestinal sialylation in neonatal mice. Glycobiology 2022; 32:414-428. [PMID: 35157771 PMCID: PMC9022908 DOI: 10.1093/glycob/cwac005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/10/2022] [Accepted: 01/30/2022] [Indexed: 11/14/2022] Open
Abstract
Interactions between the neonate host and its gut microbiome are central to the development of a healthy immune system. However, the mechanisms by which animals alter early colonization of microbiota for their benefit remain unclear. Here, we investigated the role of early-life expression of the α2,6-sialyltransferase ST6GAL1 in microbiome phylogeny and mucosal immunity. Fecal, upper respiratory, and oral microbiomes of pups expressing or lacking St6gal1 were analyzed by 16S rRNA sequencing. At weaning, the fecal microbiome of St6gal1-KO mice had reduced Clostridiodes, Coprobacillus, and Adlercreutzia, but increased Helicobacter and Bilophila. Pooled fecal microbiomes from syngeneic donors were transferred to antibiotic-treated wild-type mice, before analysis of recipient mucosal immune responses by flow cytometry, RT-qPCR, microscopy, and ELISA. Transfer of St6gal1-KO microbiome induced a mucosal Th17 response, with expression of T-bet and IL-17, and IL-22-dependent gut lengthening. Early life intestinal sialylation was characterized by RT-qPCR, immunoblot, microscopy, and sialyltransferase enzyme assays in genetic mouse models at rest or with glucocorticoid receptor modulators. St6gal1 expression was greatest in the duodenum, where it was mediated by the P1 promoter and efficiently inhibited by dexamethasone. Our data show that the inability to produce α2,6-sialyl ligands contributes to microbiome-dependent Th17 inflammation, highlighting a pathway by which the intestinal glycosylation regulates mucosal immunity.
Collapse
Affiliation(s)
- Eric E Irons
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Valerie L Andersen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| | - Joseph T Y Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, United States
| |
Collapse
|
3
|
Chen T, Oh S, Gregory S, Shen X, Diehl AM. Single-cell omics analysis reveals functional diversification of hepatocytes during liver regeneration. JCI Insight 2020. [DOI: 10.1172/jci.insight.141024 33208554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
4
|
Chen T, Oh S, Gregory S, Shen X, Diehl AM. Single-cell omics analysis reveals functional diversification of hepatocytes during liver regeneration. JCI Insight 2020; 5:141024. [PMID: 33208554 PMCID: PMC7710279 DOI: 10.1172/jci.insight.141024] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/08/2020] [Indexed: 01/07/2023] Open
Abstract
Adult liver has enormous regenerative capacity; it can regenerate after losing two-thirds of its mass while sustaining essential metabolic functions. How the liver balances dual demands for increased proliferative activity with maintenance of organ function is unknown but essential to prevent liver failure. Using partial hepatectomy (PHx) in mice to model liver regeneration, we integrated single-cell RNA- and ATAC-Seq to map state transitions in approximately 13,000 hepatocytes at single-cell resolution as livers regenerated, and validated key findings with IHC, to uncover how the organ regenerates hepatocytes while simultaneously fulfilling its vital tissue-specific functions. After PHx, hepatocytes rapidly and transiently diversified into multiple distinct populations with distinct functional bifurcation: some retained the chromatin landscapes and transcriptomes of hepatocytes in undamaged adult livers, whereas others transitioned to acquire chromatin landscapes and transcriptomes of fetal hepatocytes. Injury-related signaling pathways known to be critical for regeneration were activated in transitioning hepatocytes, and the most fetal-like hepatocytes exhibited chromatin landscapes that were enriched with transcription factors regulated by those pathways.
Collapse
Affiliation(s)
- Tianyi Chen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Simon Gregory
- Department of Neurology, Duke University, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA
| | - Anna Mae Diehl
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Medicine and
| |
Collapse
|
5
|
Ding C, Tang W, Wu H, Fan X, Luo J, Feng J, Wen K, Wu G. The PEAK1-PPP1R12B axis inhibits tumor growth and metastasis by regulating Grb2/PI3K/Akt signalling in colorectal cancer. Cancer Lett 2018; 442:383-395. [PMID: 30472186 DOI: 10.1016/j.canlet.2018.11.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/28/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
Pseudopodium enriched atypical kinase 1 (PEAK1), a novel non-receptor tyrosine kinase, was recently implicated in cancer pathogenesis. However, its functional role in colorectal cancer (CRC) is not well known. Herein, we demonstrated that PEAK1 was frequently downregulated in CRC and significantly associated with tumor size, differentiation status, metastasis, and clinical stage. PEAK1 overexpression suppressed CRC cell growth, invasion, and metastasis in vitro and in vivo, whereas knockout had the opposite effects. Further evaluation revealed that PEAK1 expression was positively correlated with protein phosphatase 1 regulatory subunit 12B (PPP1R12B) in CRC cell lines and clinical tissues, and this protein was found to suppress activation of the Grb2/PI3K/Akt pathway. Moreover, PPP1R12B knockdown markedly abrogated PEAK1-mediated tumor suppressive effects, whereas its upregulation recapitulated the effects of PEAK1 knockout on cell behaviours and the activation of signalling. Mechanistically, PI3K and Akt inhibitors reversed impaired the effect of PEAK1 function on cell proliferation, migration, and invasion. Our results provide compelling evidence that the PEAK1-PPP1R12B axis inhibits colorectal tumorigenesis and metastasis through deactivation of the Grb2/PI3K/Akt pathway, which might provide a novel therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Chenbo Ding
- Medical School of Southeast University, Nanjing, China; Center of Clinical Laboratory Medicine, The Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Wendong Tang
- Medical School of Southeast University, Nanjing, China
| | - Hailu Wu
- Medical School of Southeast University, Nanjing, China; Department of Gastroenterology, The Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Xiaobo Fan
- Medical School of Southeast University, Nanjing, China
| | - Junmin Luo
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Jihong Feng
- Department of Oncology, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kunming Wen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guoqiu Wu
- Medical School of Southeast University, Nanjing, China; Center of Clinical Laboratory Medicine, The Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
6
|
Irudayaswamy A, Muthiah M, Zhou L, Hung H, Jumat NHB, Haque J, Teoh N, Farrell G, Riehle KJ, Lin JS, Su LL, Chan JK, Choolani M, Wong PC, Wee A, Lim SG, Campbell J, Fausto N, Dan YY. Long-Term Fate of Human Fetal Liver Progenitor Cells Transplanted in Injured Mouse Livers. Stem Cells 2017; 36:103-113. [PMID: 28960647 DOI: 10.1002/stem.2710] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 08/25/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022]
Abstract
Liver progenitor cells have the potential to repair and regenerate a diseased liver. The success of any translational efforts, however, hinges on thorough understanding of the fate of these cells after transplant, especially in terms of long-term safety and efficacy. Here, we report transplantation of a liver progenitor population isolated from human fetal livers into immune-permissive mice with follow-up up to 36 weeks after transplant. We found that human progenitor cells engraft and differentiate into functional human hepatocytes in the mouse, producing albumin, alpha-1-antitrypsin, and glycogen. They create tight junctions with mouse hepatocytes, with no evidence of cell fusion. Interestingly, they also differentiate into functional endothelial cell and bile duct cells. Transplantation of progenitor cells abrogated carbon tetrachloride-induced fibrosis in recipient mice, with downregulation of procollagen and anti-smooth muscle actin. Paradoxically, the degree of engraftment of human hepatocytes correlated negatively with the anti-fibrotic effect. Progenitor cell expansion was most prominent in cirrhotic animals, and correlated with transcript levels of pro-fibrotic genes. Animals that had resolution of fibrosis had quiescent native progenitor cells in their livers. No evidence of neoplasia was observed, even up to 9 months after transplantation. Human fetal liver progenitor cells successfully attenuate liver fibrosis in mice. They are activated in the setting of liver injury, but become quiescent when injury resolves, mimicking the behavior of de novo progenitor cells. Our data suggest that liver progenitor cells transplanted into injured livers maintain a functional role in the repair and regeneration of the liver. Stem Cells 2018;36:103-113.
Collapse
Affiliation(s)
| | - Mark Muthiah
- Department of Medicine, National University Singapore, Singapore.,Division of Gastroenterology and Hepatology, National University Hospital. National University Health System, Singapore
| | - Lei Zhou
- Department of Medicine, National University Singapore, Singapore
| | - Hau Hung
- Department of Medicine, National University Singapore, Singapore
| | | | - Jamil Haque
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Narcissus Teoh
- Department of Medicine, Australian National University, Canberra, Australia
| | - Geoffrey Farrell
- Department of Medicine, Australian National University, Canberra, Australia
| | - Kimberly J Riehle
- Department of Pathology, University of Washington, Seattle, Washington, USA.,Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Jaymie Siqi Lin
- Department of Medicine, National University Singapore, Singapore
| | - Lin Lin Su
- Department of Obstetrics and Gynecology, National University Singapore, Singapore
| | - Jerry Ky Chan
- Department of Obstetrics and Gynecology, National University Singapore, Singapore.,Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynecology, National University Singapore, Singapore
| | - Peng Cheang Wong
- Department of Obstetrics and Gynecology, National University Singapore, Singapore
| | - Aileen Wee
- Department of Pathology, National University Singapore, Singapore
| | - Seng Gee Lim
- Department of Medicine, National University Singapore, Singapore.,Division of Gastroenterology and Hepatology, National University Hospital. National University Health System, Singapore
| | - Jean Campbell
- Clinical Research Divison, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nelson Fausto
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Yock Young Dan
- Department of Medicine, National University Singapore, Singapore.,Division of Gastroenterology and Hepatology, National University Hospital. National University Health System, Singapore.,Cancer Science Institute, National University Singapore, Singapore.,Genome Institute Singapore, ASTAR, Singapore
| |
Collapse
|
7
|
Li J, Wu H, Li W, Yin L, Guo S, Xu X, Ouyang Y, Zhao Z, Liu S, Tian Y, Tian Z, Ju J, Ni B, Wang H. Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-κB signaling. Oncogene 2016; 35:5501-5514. [PMID: 27065335 PMCID: PMC5078861 DOI: 10.1038/onc.2016.90] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 01/27/2016] [Accepted: 02/19/2016] [Indexed: 02/06/2023]
Abstract
The aberrant expression of microRNAs (miRNAs) has emerged as an important hallmark of cancer. However, the molecular mechanisms underlying the changes in miRNA expression remain unclear. In this study, we discovered a novel epigenetic mechanism of miR-506 regulation and investigated its functional significance in pancreatic cancer. Sequencing analysis revealed that the miR-506 promoter is highly methylated in pancreatic cancer tissues compared with non-cancerous tissues. Reduced miR-506 expression was significantly associated with clinical stage, pathologic tumor status, distant metastasis and decreased survival of pancreatic cancer patients. miR-506 inhibited cell proliferation, induced cell cycle arrest at the G1/S transition and enhanced apoptosis and chemosensitivity of pancreatic cancer cells. Furthermore, we identified sphingosine kinase 1 (SPHK1) as a novel target of miR-506, the expression of which inhibited the SPHK1/Akt/NF-κB signaling pathway, which is activated in pancreatic cancer. High SPHK1 expression was significantly associated with poor survival in a large cohort of pancreatic cancer specimens. Our data suggest that miR-506 acts as a tumor suppressor miRNA and is epigenetically silenced in pancreatic cancer. The newly identified miR-506/SPHK1 axis represents a novel therapeutic strategy for future pancreatic cancer treatment.
Collapse
Affiliation(s)
- J Li
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - H Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - W Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - L Yin
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - S Guo
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - X Xu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Y Ouyang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Z Zhao
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - S Liu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Y Tian
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - Z Tian
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
| | - J Ju
- Translational Research Laboratory, Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - B Ni
- Institute of Immunology PLA, Third Military Medical University, Chongqing, China
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - H Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
Baranov VS, Ivaschenko TE, Yarmolinskaya MI. Comparative systems genetics view of endometriosis and uterine leiomyoma: Two sides of the same coin? Syst Biol Reprod Med 2016; 62:93-105. [PMID: 26828864 DOI: 10.3109/19396368.2015.1123325] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Endometriosis (EM) and uterine leiomyoma (UL) are two most frequent benign tumors of monoclonal origin affecting about 30% of all women in their reproductive age. Modern molecular technologies have made a tremendous impact in understanding both disorders. Here is the first comparative analysis of molecular mechanisms underlying development of EM and UL as it looks from the platform of systems genetics. Similarities and differences of EM and UL at their incipient stages are enlightened with special emphasis on their gene networks, gene expression, and epigenetic regulation, of pathologic development. The analysis substantiates a new hypothesis postulating tumors as outgrowths of the stem cells with mesenchymal commitment lineage (mSC) which migrate from the endometrium/myometrium junctional zone of the uterus. Comparative analysis has revealed basic similarities of molecular pathogenesis of EM and UL suggesting molecular syntropy of both disorders. Peculiarities of the epigenetic landscape determining development of mSC may explain the existence of different clinical forms of EM and UL as well as their unique clinical manifestation. Some perspectives for practical and scientific application in EM and UL studies of this new hypothesis are outlined.
Collapse
Affiliation(s)
- Vladislav S Baranov
- a D.O. Ott Research Institute of Obstetrics , Gynecology and Reproductology , Saint-Petersburg , Russian Federation
| | - Tatyana E Ivaschenko
- a D.O. Ott Research Institute of Obstetrics , Gynecology and Reproductology , Saint-Petersburg , Russian Federation
| | - Maria I Yarmolinskaya
- a D.O. Ott Research Institute of Obstetrics , Gynecology and Reproductology , Saint-Petersburg , Russian Federation
| |
Collapse
|
9
|
Revisiting Epithelial-to-Mesenchymal Transition in Liver Fibrosis: Clues for a Better Understanding of the "Reactive" Biliary Epithelial Phenotype. Stem Cells Int 2016; 2016:2953727. [PMID: 26880950 PMCID: PMC4736590 DOI: 10.1155/2016/2953727] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/20/2015] [Indexed: 12/27/2022] Open
Abstract
Whether liver epithelial cells contribute to the development of hepatic scarring by undergoing epithelial-to-mesenchymal transition (EMT) is a controversial issue. Herein, we revisit the concept of EMT in cholangiopathies, a group of severe hepatic disorders primarily targeting the bile duct epithelial cell (cholangiocyte), leading to progressive portal fibrosis, the main determinant of liver disease progression. Unfortunately, therapies able to halt this process are currently lacking. In cholangiopathies, fibrogenesis is part of ductular reaction, a reparative complex involving epithelial, mesenchymal, and inflammatory cells. Ductular reactive cells (DRC) are cholangiocytes derived from the activation of the hepatic progenitor cell compartment. These cells are arranged into irregular strings and express a “reactive” phenotype, which enables them to extensively crosstalk with the other components of ductular reaction. We will first discuss EMT in liver morphogenesis and then highlight how some of these developmental programs are partly reactivated in DRC. Evidence for “bona fide” EMT changes in cholangiocytes is lacking, but expression of some mesenchymal markers represents a fundamental repair mechanism in response to chronic biliary damage with potential harmful fibrogenetic effects. Understanding microenvironmental cues and signaling perturbations promoting these changes in DRC may help to identify potential targets for new antifibrotic therapies in cholangiopathies.
Collapse
|
10
|
Brivio S, Cadamuro M, Fabris L, Strazzabosco M. Epithelial-to-Mesenchymal Transition and Cancer Invasiveness: What Can We Learn from Cholangiocarcinoma? J Clin Med 2015; 4:2028-41. [PMID: 26703747 PMCID: PMC4693158 DOI: 10.3390/jcm4121958] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/04/2015] [Accepted: 12/14/2015] [Indexed: 12/11/2022] Open
Abstract
In addition to its well-established role in embryo development, epithelial-to-mesenchymal transition (EMT) has been proposed as a general mechanism favoring tumor metastatization in several epithelial malignancies. Herein, we review the topic of EMT in cholangiocarcinoma (CCA), a primary liver cancer arising from the epithelial cells lining the bile ducts (cholangiocytes) and characterized by an abundant stromal reaction. CCA carries a dismal prognosis, owing to a pronounced invasiveness and scarce therapeutic opportunities. In CCA, several reports indicate that cancer cells acquire a number of EMT biomarkers and functions. These phenotypic changes are likely induced by both autocrine and paracrine signals released in the tumor microenvironment (cytokines, growth factors, morphogens) and intracellular stimuli (microRNAs, oncogenes, tumor suppressor genes) variably associated with specific disease mechanisms, including chronic inflammation and hypoxia. Nevertheless, evidence supporting a complete EMT of neoplastic cholangiocytes into stromal cells is lacking, and the gain of EMT-like changes by CCA cells rather reflects a shift towards an enhanced pro-invasive phenotype, likely induced by the tumor stroma. This concept may help to identify new biomarkers of early metastatic behavior along with potential therapeutic targets.
Collapse
Affiliation(s)
- Simone Brivio
- School of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900 Monza, Italy.
| | - Massimiliano Cadamuro
- School of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900 Monza, Italy.
- Department of Molecular Medicine, University of Padua School of Medicine, Viale Colombo 3, 35131 Padua, Italy.
| | - Luca Fabris
- Department of Molecular Medicine, University of Padua School of Medicine, Viale Colombo 3, 35131 Padua, Italy.
- Liver Center, Section of Digestive Diseases, Yale University, TAC Building, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Mario Strazzabosco
- School of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900 Monza, Italy.
- Liver Center, Section of Digestive Diseases, Yale University, TAC Building, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
11
|
Identification of Pathways Mediating Growth Differentiation Factor5-Induced Tenogenic Differentiation in Human Bone Marrow Stromal Cells. PLoS One 2015; 10:e0140869. [PMID: 26528540 PMCID: PMC4631504 DOI: 10.1371/journal.pone.0140869] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
To date, the molecular signalling mechanisms which regulate growth factors-induced MSCs tenogenic differentiation remain largely unknown. Therefore, a study to determine the global gene expression profile of tenogenic differentiation in human bone marrow stromal cells (hMSCs) using growth differentiation factor 5 (GDF5) was conducted. Microarray analyses were conducted on hMSCs cultures supplemented with 100 ng/ml of GDF5 and compared to undifferentiated hMSCs and adult tenocytes. Results of QuantiGene® Plex assay support the use and interpretation of the inferred gene expression profiles and pathways information. From the 27,216 genes assessed, 873 genes (3.21% of the overall human transcriptome) were significantly altered during the tenogenic differentiation process (corrected p<0.05). The genes identified as potentially associated with tenogenic differentiation were ARHGAP29, CCL2, integrin alpha 8 and neurofilament medium polypeptides. These genes, were mainly associated with cytoskeleton reorganization (stress fibers formation) signaling. Pathway analysis demonstrated the potential molecular pathways involved in tenogenic differentiation were: cytoskeleton reorganization related i.e. keratin filament signaling and activin A signaling; cell adhesion related i.e. chemokine and adhesion signaling; and extracellular matrix related i.e. arachidonic acid production signaling. Further investigation using atomic force microscopy and confocal laser scanning microscopy demonstrated apparent cytoskeleton reorganization in GDF5-induced hMSCs suggesting that cytoskeleton reorganization signaling is an important event involved in tenogenic differentiation. Besides, a reduced nucleostemin expression observed suggested a lower cell proliferation rate in hMSCs undergoing tenogenic differentiation. Understanding and elucidating the tenogenic differentiation signalling pathways are important for future optimization of tenogenic hMSCs for functional tendon cell-based therapy and tissue engineering.
Collapse
|
12
|
Liu D, Yovchev MI, Zhang J, Alfieri AA, Tchaikovskaya T, Laconi E, Dabeva MD. Identification and characterization of mesenchymal-epithelial progenitor-like cells in normal and injured rat liver. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:110-28. [PMID: 25447047 DOI: 10.1016/j.ajpath.2014.08.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 08/22/2014] [Accepted: 08/27/2014] [Indexed: 01/07/2023]
Abstract
In normal rat liver, thymocyte antigen 1 (Thy1) is expressed in fibroblasts/myofibroblasts and in some blood progenitor cells. Thy1-expressing cells also accumulate in the liver during impaired liver regeneration. The origin and nature of these cells are not well understood. By using RT-PCR analysis and immunofluorescence microscopy, we describe the presence of rare Thy1(+) cells in the liver lobule of normal animals, occasionally forming small collections of up to 20 cells. These cells constitute a small portion (1.7% to 1.8%) of nonparenchymal cells and reveal a mixed mesenchymal-epithelial phenotype, expressing E-cadherin, cytokeratin 18, and desmin. The most potent mitogens for mesenchymal-epithelial Thy1(+) cells in vitro are the inflammatory cytokines interferon γ, IL-1, and platelet-derived growth factor-BB, which are not produced by Thy1(+) cells. Thy1(+) cells express all typical mesenchymal stem cell and hepatic progenitor cell markers and produce growth factor and cytokine mRNA (Hgf, Il6, Tgfa, and Tweak) for proteins that maintain oval cell growth and differentiation. Under appropriate conditions, mesenchymal-epithelial cells differentiate in vitro into hepatocyte-like cells. In this study, we show that the adult rat liver harbors a small pool of endogenous mesenchymal-epithelial cells not recognized previously. In the quiescent state, these cells express both mesenchymal and epithelial cell markers. They behave like hepatic stem cells/progenitors with dual phenotype, exhibiting high plasticity and long-lasting proliferative activity.
Collapse
Affiliation(s)
- Daqing Liu
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Mladen I Yovchev
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jinghang Zhang
- Flow Cytometry Core Facility, Albert Einstein College of Medicine, Bronx, New York
| | - Alan A Alfieri
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Tatyana Tchaikovskaya
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Ezio Laconi
- Section of Experimental Pathology, Department of Sciences and Biomedical Technology, University of Cagliari, Cagliari, Italy
| | - Mariana D Dabeva
- Department of Medicine, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
13
|
Chang JH, Au HK, Lee WC, Chi CC, Ling TY, Wang LM, Kao SH, Huang YH, Tzeng CR. Expression of the pluripotent transcription factor OCT4 promotes cell migration in endometriosis. Fertil Steril 2013; 99:1332-1339.e5. [PMID: 23290742 DOI: 10.1016/j.fertnstert.2012.11.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 11/09/2012] [Accepted: 11/16/2012] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To identify the impact of the pluripotent transcription factor OCT4 in endometrial cell migration and endometriosis. DESIGN The OCT4 expression and cell migration study. SETTING Research institution and reproductive medical clinic. PATIENT(S) Nine subjects with normal endometrium, 3 subjects with normal myometrium, 36 patients with hyperplastic endometrium, and 58 patients with endometriosis. INTERVENTION(S) The expression of OCT4 messenger RNA in normal endometrium, normal myometrium, hyperplastic endometrium, and ectopic endometriotic tissues was analyzed using reverse transcription and quantitative real-time polymerase chain reaction (PCR). The effect of OCT4 expression on the migration activity of the endometrial cells was examined. MAIN OUTCOME MEASURE(S) Reverse transcription and quantitative real-time PCR, Western blotting, and wound closure and transwell assays. RESULT(S) The expression of OCT4 and NANOG messenger RNA was significantly higher in ectopic endometriotic tissues, compared with that of the normal endometrium, the normal myometrium, and the hyperplastic endometrium. The level of OCT4 messenger RNA in endometriotic tissues was positively correlated with the expression of genes associated with cell migration. Overexpression of the OCT4 protein in primary human endometriotic stromal cells and human RL95-2 and HEC1A endometrial carcinoma cell lines resulted in decreased levels of E-CADHERIN, the increased expression of the VIMENTIN, TWIST, and SLUG proteins, and an increase in the migration activity of endometrial cells in transwell and wound closure assays. CONCLUSION(S) The transcription of the OCT4 gene is significantly up-regulated in human ectopic endometriotic tissues. The expression of OCT4 may contribute to the pathology of ectopic endometrial growth by stimulating the migration activity of endometrial cells.
Collapse
Affiliation(s)
- Jui-Hung Chang
- Department of Biochemistry, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Wang M, Hada M, Huff J, Pluth JM, Anderson J, O'Neill P, Cucinotta FA. Heavy ions can enhance TGFβ mediated epithelial to mesenchymal transition. JOURNAL OF RADIATION RESEARCH 2012; 53:51-57. [PMID: 22302045 DOI: 10.1269/jrr.11121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
TGFβ is a key modulator of the Epithelial-Mesenchymal Transition (EMT), a process important in cancer progression and metastasis, which leads to the suppression of epithelial genes and expression of mesenchymal proteins. Ionizing radiation was found to specifically induce expression of the TGF-β1 isoform, which can modulate late post-radiation changes and increase the risk of tumor development and metastasis. Interactions between TGFβ induced EMT and DNA damage responses have not been fully elucidated, particularly at low doses and following different radiation quality exposures. Further characterization of the relationship between radiation quality, EMT and cancer development is warranted. We investigated whether space radiation induced TGFβ dependent EMT, using hTERT immortalized human esophageal epithelial cells (EPC2-hTERT) and non-transformed mink lung epithelial cells (Mv1Lu). We have observed morphologic and molecular alterations in EPC2 and Mv1Lu cells consistent with EMT after pre-treatment with TGFβ1. This effect could be efficiently inhibited in both cell lines by the use of a TGFβRI inhibitor. High-energy silicon or iron nuclei were each able to cause a mild induction of EMT, with the inclusion of TGFβ1 inducing a greatly enhanced EMT phenotype even when cells were irradiated with doses as low as 0.1 Gy. A further enhancement of EMT was achieved at a higher dose of 2 Gy. TGFβRI inhibitor was able to reverse the EMT induced by the combination of TGFβ1 and radiation. These studies indicate that heavy ions, even at a low dose, may trigger the process of TGFβ1-induced EMT, and suggest further studies are needed to determine whether the chronic exposures received in space may potentiate this process in astronauts, leading to an increased risk of cancer.
Collapse
Affiliation(s)
- Minli Wang
- USRA, Division of Life Sciences, Houston, TX 77058, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Ogunwobi OO, Liu C. Therapeutic and prognostic importance of epithelial-mesenchymal transition in liver cancers: insights from experimental models. Crit Rev Oncol Hematol 2011; 83:319-28. [PMID: 22178416 DOI: 10.1016/j.critrevonc.2011.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 11/08/2011] [Accepted: 11/23/2011] [Indexed: 12/22/2022] Open
Abstract
Hepatocellular carcinoma (HCC) and hepatic metastases of colon cancers are malignant conditions of the liver that cause significant morbidity and mortality worldwide. Experimental models suggest that both conditions are characterized by epithelial-mesenchymal transition (EMT). Whilst ongoing research efforts aim to definitively clarify its role in human cancer patients, data from experimental models have unraveled a number of potential therapeutic targets as well as markers of prognostic importance. This area of research is generating intense interest amongst both basic scientists and clinicians. Some questions have been answered, but many important issues remain unresolved. We expect that in the near future, studies of human tissues can definitively clarify the role of EMT in the development and progression of human malignant diseases of the liver and that further studies can be carried out to determine how best to target aspects of the process for the treatment of patients with hepatocellular carcinoma and hepatic metastases of colon cancers.
Collapse
Affiliation(s)
- Olorunseun O Ogunwobi
- Department of Pathology, Immunology and Laboratory Medicine and Shands Cancer Center, University of Florida, Gainesville, FL, USA.
| | | |
Collapse
|
16
|
LI Y, WANG XY, WANG LJ, XU T, LU XY, CAI DQ, GENG JG, YANG XS. PTEN impedes EMT during chick embryo gastrulation. YI CHUAN = HEREDITAS 2011; 33:613-9. [DOI: 10.3724/sp.j.1005.2011.00613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|