1
|
Wu Z, Zhang Z, Zhou S, Xie M, Liu L, Luo C, Zheng F, Qiu W, Wang Y, Zhang J. ERK1/2-dependent activity of SOX9 is required for sublytic C5b-9-induced expression of FGF1, PDGFα, and TGF-β1 in rat Thy-1 nephritis. Int Immunopharmacol 2024; 127:111372. [PMID: 38118314 DOI: 10.1016/j.intimp.2023.111372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 12/22/2023]
Abstract
Mesangial proliferative glomerulonephritis (MsPGN) and its related rat model Thy-1 nephritis (Thy-1N) are associated with C5b-9 deposition and are characterized by proliferation of glomerular mesangial cell (GMC) and expansion of extracellular matrix (ECM) expansion, alongside overexpression of multiple growth factors. Although fibroblast growth factor 1 (FGF1), platelet-derived growth factor alpha (PDGFα), and transforming growth factor beta 1 (TGF-β1) are well known for their proproliferative and profibrotic roles, the molecular mechanisms responsible for regulating the expression of these growth factors have not been thoroughly elucidated. In this study, we found that sublytic C5b-9 induction of sex-determining region Y-box 9 (SOX9) transactivated FGF1, PDGFα, and TGF-β1 genes in GMCs, resulting in a significant increase in their mRNA and protein levels. Besides, sublytic C5b-9 induction of activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylated SOX9 at serine 181 and serine 64, which enhanced SOX9's ability to transactivate FGF1, PDGFα, and TGF-β1 genes in GMCs. Furthermore, we demonstrated that inhibiting ERK1/2 activation or silencing either ERK1/2 or SOX9 gene led to reduced SOX9 phosphorylation, decreased generation of FGF1, PDGFα, and TGF-β1, and ameliorated glomerular injury in rat Thy-1N. Overall, these findings suggest that expression of FGF1, PDGFα, and TGF-β1 is promoted by ERK1/2-mediated phosphorylation of SOX9, which may provide a valuable insight into the pathogenesis of MsPGN and offer a potential target for the development of novel treatment strategies for MsPGN.
Collapse
Affiliation(s)
- Zhijiao Wu
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhiwei Zhang
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Sicheng Zhou
- School of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mengxiao Xie
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Longfei Liu
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Can Luo
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feixiang Zheng
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Wen Qiu
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, China; National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, China
| | - Yingwei Wang
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, China; National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Department of Immunology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China; Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, China; National Health Commission Key Laboratory of Antibody Techniques, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Nitta Y, Fujii T, Uchiyama T, Sugimoto A, Nishikawa T, Takeda M, Miyake M, Shimada K, Fujimoto K. Overexpression of MicroRNA-138 Affects the Proliferation and Invasion of Urothelial Carcinoma Cells by Suppressing SOX9 Expression. Biomedicines 2023; 11:3064. [PMID: 38002064 PMCID: PMC10669193 DOI: 10.3390/biomedicines11113064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
SRY-box transcription factor 9 (SOX9) is important for sexual differentiation, chondrogenic differentiation, and cell proliferation in cancer. It acts as a target molecule of microRNA (miR)-138 in various tumors and is associated with tumor development and growth. In this study, we analyzed the functions of miR-138 and SOX9 in urothelial carcinoma. SOX9 was highly expressed in invasive urothelial carcinoma tissues. miR-138 precursor transfection of T24 and UMUC2 cells significantly decreased SOX9 expression, indicating that SOX9 is a miR-138 target in urothelial carcinoma. Moreover, miR-138 precursor or SOX9 small interfering RNA (siRNA) transfection decreased the proliferation of urothelial carcinoma cell lines. To further confirm that miR-138-SOX9 signaling is involved in cell proliferation and invasion, urothelial carcinoma cells were transfected with the miR-138 precursor or SOX9 siRNA. This transfection reduced the proliferation and invasion of cells via the promotion of autophagy and apoptosis and G0/G1 cell cycle arrest. These results suggest that miR-138-SOX9 signaling modulates the growth and invasive potential of urothelial carcinoma cells.
Collapse
Affiliation(s)
- Yuji Nitta
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
- Division of Fostering Required Medical Human Resources, Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan
| | - Tomoko Uchiyama
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| | - Aya Sugimoto
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| | - Takeshi Nishikawa
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
- Department of Central Clinical Laboratory, Nara Medical University Hospital, Nara 634-8521, Japan
| | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| | - Makito Miyake
- Department of Urology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| | - Keiji Shimada
- Department of Diagnostic Pathology, Nara City Hospital, Nara 630-8305, Japan
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University School of Medicine, Nara 634-8521, Japan
| |
Collapse
|
3
|
Baddela VS, Michaelis M, Tao X, Koczan D, Vanselow J. ERK1/2-SOX9/FOXL2 axis regulates ovarian steroidogenesis and favors the follicular-luteal transition. Life Sci Alliance 2023; 6:e202302100. [PMID: 37532283 PMCID: PMC10397509 DOI: 10.26508/lsa.202302100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Estradiol and progesterone are the primary sex steroids produced by the ovary. Upon luteinizing hormone surge, estradiol-producing granulosa cells convert into progesterone-producing cells and eventually become large luteal cells of the corpus luteum. Signaling pathways and transcription factors involved in the cessation of estradiol and simultaneous stimulation of progesterone production in granulosa cells are not clearly understood. Here, we decipher that phosphorylated ERK1/2 regulates granulosa cell steroidogenesis by inhibiting estradiol and inducing progesterone production. Down-regulation of transcription factor FOXL2 and up-regulation of SOX9 by ERK underpin its differential steroidogenic function. Interestingly, the incidence of SOX9 is largely uncovered in ovarian cells and is found to regulate FOXL2 along with CYP19A1 and STAR genes, encoding rate-limiting enzymes of steroidogenesis, in cultured granulosa cells. We propose that the novel ERK1/2-SOX9/FOXL2 axis in granulosa cells is a critical regulator of ovarian steroidogenesis and may be considered when addressing pathophysiologies associated with inappropriate steroid production and infertility in humans and animals.
Collapse
Affiliation(s)
- Vijay Simha Baddela
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Marten Michaelis
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Xuelian Tao
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Dirk Koczan
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - Jens Vanselow
- Institute of Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
4
|
Zhong H, Lu W, Tang Y, Wiel C, Wei Y, Cao J, Riedlinger G, Papagiannakopoulos T, Guo JY, Bergo MO, Kang Y, Ganesan S, Sabaawy HE, Pine SR. SOX9 drives KRAS-induced lung adenocarcinoma progression and suppresses anti-tumor immunity. Oncogene 2023:10.1038/s41388-023-02715-5. [PMID: 37258742 DOI: 10.1038/s41388-023-02715-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023]
Abstract
The SOX9 transcription factor ensures proper tissue development and homeostasis and has been implicated in promoting tumor progression. However, the role of SOX9 as a driver of lung adenocarcinoma (LUAD), or any cancer, remains unclear. Using CRISPR/Cas9 and Cre-LoxP gene knockout approaches in the KrasG12D-driven mouse LUAD model, we found that loss of Sox9 significantly reduces lung tumor development, burden and progression, contributing to significantly longer overall survival. SOX9 consistently drove organoid growth in vitro, but SOX9-promoted tumor growth was significantly attenuated in immunocompromised mice compared to syngeneic mice. We demonstrate that SOX9 suppresses immune cell infiltration and functionally suppresses tumor associated CD8+ T, natural killer and dendritic cells. These data were validated by flow cytometry, gene expression, RT-qPCR, and immunohistochemistry analyses in KrasG12D-driven murine LUAD, then confirmed by interrogating bulk and single-cell gene expression repertoires and immunohistochemistry in human LUAD. Notably, SOX9 significantly elevates collagen-related gene expression and substantially increases collagen fibers. We propose that SOX9 increases tumor stiffness and inhibits tumor-infiltrating dendritic cells, thereby suppressing CD8+ T cell and NK cell infiltration and activity. Thus, SOX9 drives KrasG12D-driven lung tumor progression and inhibits anti-tumor immunity at least partly by modulating the tumor microenvironment.
Collapse
Affiliation(s)
- Hua Zhong
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Wen Lu
- Howard Hughes Medical Institute, Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Department of Medicine, Department of Microbiology and Immunology, University of California, San Francisco, CA, 94143-0795, USA
| | - Yong Tang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Clotilde Wiel
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, 405 30, Gothenburg, Sweden
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Jian Cao
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Gregory Riedlinger
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Thales Papagiannakopoulos
- Perlmutter NYU Cancer Center, Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Jessie Yanxiang Guo
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA
| | - Martin O Bergo
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Hatim E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sharon R Pine
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
5
|
Bhatia R, Siddiqui JA, Ganguly K, Thompson CM, Cannon A, Aithal A, Perumal N, Maurya SK, Li X, Cox JL, Gurumurthy CB, Rachagani S, Jain M, Nasser MW, Batra SK, Kumar S. Muc4 loss mitigates epidermal growth factor receptor activity essential for PDAC tumorigenesis. Oncogene 2023; 42:759-770. [PMID: 36624189 PMCID: PMC10198580 DOI: 10.1038/s41388-022-02587-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023]
Abstract
Mucin4 (MUC4) appears early during pancreatic intraepithelial neoplasia-1 (PanIN1), coinciding with the expression of epidermal growth factor receptor-1 (EGFR). The EGFR signaling is required for the onset of Kras-driven pancreatic ductal adenocarcinoma (PDAC); however, the players and mechanisms involved in sustained EGFR signaling in early PanIN lesions remain elusive. We generated a unique Esai-CRISPR-based Muc4 conditional knockout murine model to evaluate its effect on PDAC pathology. The Muc4 depletion in the autochthonous murine model carrying K-ras and p53 mutations (K-rasG12D; TP53R172H; Pdx-1cre, KPC) to generate the KPCM4-/- murine model showed a significant delay in the PanIN lesion formation with a significant reduction (p < 0.01) in EGFR (Y1068) and ERK1/2 (T202/Y204) phosphorylation. Further, a significant decrease (p < 0.01) in Sox9 expression in PanIN lesions of KPCM4-/- mice suggested the impairment of acinar-to-ductal metaplasia in Muc4-depleted cells. The biochemical analyses demonstrated that MUC4, through its juxtamembrane EGF-like domains, interacts with the EGFR ectodomain, and its cytoplasmic tail prevents EGFR ubiquitination and subsequent proteasomal degradation upon ligand stimulation, leading to sustained downstream oncogenic signaling. Targeting the MUC4 and EGFR interacting interface provides a promising strategy to improve the efficacy of EGFR-targeted therapies in PDAC and other MUC4-expressing malignancies.
Collapse
Affiliation(s)
- Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jawed Akhtar Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, Omaha, NE, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Christopher M Thompson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andrew Cannon
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Naveenkumar Perumal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shailendra K Maurya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiaoqi Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, Omaha, NE, USA
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffett Cancer Center, Omaha, NE, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffett Cancer Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Morgado-Pascual JL, Suarez-Alvarez B, Marchant V, Basantes P, Tharaux PL, Ortiz A, Lopez-Larrea C, Ruiz-Ortega M, Rayego-Mateos S. Type IV Collagen and SOX9 Are Molecular Targets of BET Inhibition in Experimental Glomerulosclerosis. Int J Mol Sci 2022; 24:486. [PMID: 36613933 PMCID: PMC9820124 DOI: 10.3390/ijms24010486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/14/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Progressive glomerulonephritis (GN) is characterized by an excessive accumulation of extracellular (ECM) proteins, mainly type IV collagen (COLIV), in the glomerulus leading to glomerulosclerosis. The current therapeutic approach to GN is suboptimal. Epigenetic drugs could be novel therapeutic options for human disease. Among these drugs, bromodomain and extra-terminal domain (BET) inhibitors (iBETs) have shown beneficial effects in experimental kidney disease and fibrotic disorders. Sex-determining region Y-box 9 (SOX9) is a transcription factor involved in regulating proliferation, migration, and regeneration, but its role in kidney fibrosis is still unclear. We investigated whether iBETs could regulate ECM accumulation in experimental GN and evaluated the role of SOX9 in this process. For this purpose, we tested the iBET JQ1 in mice with anti-glomerular basement membrane nephritis induced by nephrotoxic serum (NTS). In NTS-injected mice, JQ1 treatment reduced glomerular ECM deposition, mainly by inhibiting glomerular COLIV accumulation and Col4a3 gene overexpression. Moreover, chromatin immunoprecipitation assays demonstrated that JQ1 inhibited the recruitment and binding of BRD4 to the Col4a3 promoter and reduced its transcription. Active SOX9 was found in the nuclei of glomerular cells of NTS-injured kidneys, mainly in COLIV-stained regions. JQ1 treatment blocked SOX9 nuclear translocation in injured kidneys. Moreover, in vitro JQ1 blocked TGF-β1-induced SOX9 activation and ECM production in cultured mesangial cells. Additionally, SOX9 gene silencing inhibited ECM production, including COLIV production. Our results demonstrated that JQ1 inhibited SOX9/COLIV, to reduce experimental glomerulosclerosis, supporting further research of iBET as a potential therapeutic option in progressive glomerulosclerosis.
Collapse
Affiliation(s)
- José Luis Morgado-Pascual
- Cellular Biology in Renal Diseases Laboratory, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba University, 14004 Cordoba, Spain
| | - Beatriz Suarez-Alvarez
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
- Translational Immunology, Principality of Asturias Health Research Institute (ISPA), Central University Hospital of Asturias, 33011 Oviedo, Spain
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
| | - Pamela Basantes
- Cellular Biology in Renal Diseases Laboratory, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Center—PARCC, INSERM, Paris Cité University, 75015 Paris, France
| | - Alberto Ortiz
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
- Division of Nephrology and Hypertension, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
| | - Carlos Lopez-Larrea
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
- Translational Immunology, Principality of Asturias Health Research Institute (ISPA), Central University Hospital of Asturias, 33011 Oviedo, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, Jiménez Díaz Foundation Health Research Institute, Autonomous University of Madrid, 28040 Madrid, Spain
- REDINREN Spain/Ricord2040, 28029 Madrid, Spain
| |
Collapse
|
7
|
Jafari NV, Rohn JL. The urothelium: a multi-faceted barrier against a harsh environment. Mucosal Immunol 2022; 15:1127-1142. [PMID: 36180582 PMCID: PMC9705259 DOI: 10.1038/s41385-022-00565-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
All mucosal surfaces must deal with the challenge of exposure to the outside world. The urothelium is a highly specialized layer of stratified epithelial cells lining the inner surface of the urinary bladder, a gruelling environment involving significant stretch forces, osmotic and hydrostatic pressures, toxic substances, and microbial invasion. The urinary bladder plays an important barrier role and allows the accommodation and expulsion of large volumes of urine without permitting urine components to diffuse across. The urothelium is made up of three cell types, basal, intermediate, and umbrella cells, whose specialized functions aid in the bladder's mission. In this review, we summarize the recent insights into urothelial structure, function, development, regeneration, and in particular the role of umbrella cells in barrier formation and maintenance. We briefly review diseases which involve the bladder and discuss current human urothelial in vitro models as a complement to traditional animal studies.
Collapse
Affiliation(s)
- Nazila V Jafari
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK.
| |
Collapse
|
8
|
Kidger AM, Saville MK, Rushworth LK, Davidson J, Stellzig J, Ono M, Kuebelsbeck LA, Janssen KP, Holzmann B, Morton JP, Sansom OJ, Caunt CJ, Keyse SM. Suppression of mutant Kirsten-RAS (KRAS G12D)-driven pancreatic carcinogenesis by dual-specificity MAP kinase phosphatases 5 and 6. Oncogene 2022; 41:2811-2823. [PMID: 35418690 PMCID: PMC9106580 DOI: 10.1038/s41388-022-02302-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 12/20/2022]
Abstract
The cytoplasmic phosphatase DUSP6 and its nuclear counterpart DUSP5 are negative regulators of RAS/ERK signalling. Here we use deletion of either Dusp5 or Dusp6 to explore the roles of these phosphatases in a murine model of KRASG12D-driven pancreatic cancer. By 56-days, loss of either DUSP5 or DUSP6 causes a significant increase in KRASG12D-driven pancreatic hyperplasia. This is accompanied by increased pancreatic acinar to ductal metaplasia (ADM) and the development of pre-neoplastic pancreatic intraepithelial neoplasia (PanINs). In contrast, by 100-days, pancreatic hyperplasia is reversed with significant atrophy of pancreatic tissue and weight loss observed in animals lacking either DUSP5 or DUSP6. On further ageing, Dusp6-/- mice display accelerated development of metastatic pancreatic ductal adenocarcinoma (PDAC), while in Dusp5-/- animals, although PDAC development is increased this process is attenuated by atrophy of pancreatic acinar tissue and severe weight loss in some animals before cancer could progress. Our data suggest that despite a common target in the ERK MAP kinase, DUSP5 and DUSP6 play partially non-redundant roles in suppressing oncogenic KRASG12D signalling, thus retarding both tumour initiation and progression. Our data suggest that loss of either DUSP5 or DUSP6, as observed in certain human tumours, including the pancreas, could promote carcinogenesis.
Collapse
Affiliation(s)
- Andrew M Kidger
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Mark K Saville
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Linda K Rushworth
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Jane Davidson
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Julia Stellzig
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Motoharu Ono
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Ludwig A Kuebelsbeck
- Department of Surgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Holzmann
- Department of Surgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jennifer P Morton
- Institute of Cancer Sciences, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Owen J Sansom
- Institute of Cancer Sciences, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Christopher J Caunt
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Stephen M Keyse
- Stress Response Laboratory, Jacqui Wood Cancer Centre, Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
9
|
Lam KK, Tang CL, Tan E, Wong SH, Cheah PY. KRAS mutation-independent downregulation of MAPK/PI3K signaling in colorectal cancer. Mol Oncol 2021; 16:1171-1183. [PMID: 34919787 PMCID: PMC8895447 DOI: 10.1002/1878-0261.13163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022] Open
Abstract
KRAS is a gatekeeper gene in human colorectal tumorigenesis. KRAS is ‘undruggable’; hence, efforts have been diverted to inhibit downstream RAF/MEK/ERK and PI3K/Akt signaling. Nevertheless, none of these inhibitors has progressed to clinical use despite extensive trials. We examined levels of phospho‐ERK1/2(T202/Y204) and phospho‐Akt1/2/3(S473) in human colorectal tumor compared to matched mucosa with semi‐quantitative near‐infrared western blot and confocal fluorescence immunohistochemistry imaging. Surprisingly, 75.5% (25/33) of tumors had lower or equivalent phospho‐ERK1/2 and 96.9% (31/32) of tumors had lower phospho‐Akt1/2/3 compared to matched mucosa, irrespective of KRAS mutation status. In contrast, we discovered KRAS‐dependent SOX9 upregulation in 28 of the 31 (90.3%) tumors. These observations were substantiated by analysis of the public domain transcriptomics The Cancer Genome Atlas (TCGA) and NCBI Gene Expression Omnibus (GEO) datasets and proteomics Clinical Proteomic Tumor Analysis Consortium (CPTAC) dataset. These data suggest that RAF/MEK/ERK and PI3K/Akt signaling are unlikely to be activated in most human colorectal cancer.
Collapse
Affiliation(s)
- Kuen Kuen Lam
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | - Choong Leong Tang
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | - Emile Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | | | - Peh Yean Cheah
- Department of Colorectal Surgery, Singapore General Hospital, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore
| |
Collapse
|
10
|
Oda S, Fujisawa M, Chunning L, Ito T, Yamaguchi T, Yoshimura T, Matsukawa A. Expression of Spred2 in the urothelial tumorigenesis of the urinary bladder. PLoS One 2021; 16:e0254289. [PMID: 34818323 PMCID: PMC8612556 DOI: 10.1371/journal.pone.0254289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/08/2021] [Indexed: 11/19/2022] Open
Abstract
Aberrant activation of the Ras/Raf/ERK (extracellular-signal-regulated kinase)-MAPK (mitogen-activated protein kinase) pathway is involved in the progression of cancer, including urothelial carcinoma; but the negative regulation remains unclear. In the present study, we investigated pathological expression of Spred2 (Sprouty-related EVH1 domain-containing protein 2), a negative regulator of the Ras/Raf/ERK-MAPK pathway, and the relation to ERK activation and Ki67 index in various categories of 275 urothelial tumors obtained from clinical patients. In situ hybridization demonstrated that Spred2 mRNA was highly expressed in high-grade non-invasive papillary urothelial carcinoma (HGPUC), and the expression was decreased in carcinoma in situ (CIS) and infiltrating urothelial carcinoma (IUC). Immunohistochemically, membranous Spred2 expression, important to interact with Ras/Raf, was preferentially found in HGPUC. Interestingly, membranous Spred2 expression was decreased in CIS and IUC relative to HGPUC, while ERK activation and the expression of the cell proliferation marker Ki67 index were increased. HGPUC with membranous Spred2 expression correlated significantly with lower levels of ERK activation and Ki67 index as compared to those with negative Spred2 expression. Thus, our pathological findings suggest that Spred2 counters cancer progression in non-invasive papillary carcinoma possibly through inhibiting the Ras/Raf/ERK-MAPK pathway, but this regulatory mechanism is lost in cancers with high malignancy. Spred2 appears to be a key regulator in the progression of non-invasive bladder carcinoma.
Collapse
Affiliation(s)
- Shinsuke Oda
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masayoshi Fujisawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Li Chunning
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Japan
| | - Takahiro Yamaguchi
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- * E-mail:
| |
Collapse
|
11
|
Mori Y, Takashima S, Kanatsu-Shinohara M, Yi Z, Shinohara T. Cdc42 is required for male germline niche development in mice. Cell Rep 2021; 36:109550. [PMID: 34407418 DOI: 10.1016/j.celrep.2021.109550] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are maintained in a special microenvironment called a niche. However, much is unknown about components that constitute the niche. Here, we report that Cdc42 is essential for germline niche development. Sertoli cell-specific Cdc42-deficient mice showed normal premeiotic spermatogenesis. However, germ cells gradually disappeared during haploid cell formation and few germ cells remained in the mature testes. Spermatogonial transplantation experiments revealed a significant loss of SSCs in Cdc42-deficient testes. Moreover, Cdc42 deficiency in Sertoli cells downregulated GDNF, a critical factor for SSC maintenance. Cdc42-deficient Sertoli cells also exhibited lower nuclear MAPK1/3 staining. Inhibition of MAP2K1 or depletion of Pea15a scaffold protein downregulated GDNF expression. A screen of transcription factors revealed that Cdc42-deficient Sertoli cells downregulate DMRT1 and SOX9, both of which are critical for Sertoli cell development. These results indicate that Cdc42 is essential for niche function via MAPK1/3-dependent GDNF secretion.
Collapse
Affiliation(s)
- Yoshifumi Mori
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Seiji Takashima
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, AMED 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Zheng Yi
- Division of Experimental Hematology, Molecular Developmental Biology Graduate Program, Children's Hospital Research Foundation, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
12
|
Liu L, Ge W, Zhang Z, Li Y, Xie M, Zhao C, Yao C, Luo C, Wu Z, Wang W, Zhao D, Zhang J, Qiu W, Wang Y. Sublytic C5b-9 triggers glomerular mesangial cell proliferation via enhancing FGF1 and PDGFα gene transcription mediated by GCN5-dependent SOX9 acetylation in rat Thy-1 nephritis. FASEB J 2021; 35:e21751. [PMID: 34156114 DOI: 10.1096/fj.202002814rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022]
Abstract
Rat Thy-1 nephritis (Thy-1N) is an animal model of human mesangioproliferative glomerulonephritis (MsPGN), accompanied by glomerular mesangial cell (GMC) proliferation and extracellular matrix (ECM) deposition. Although sublytic C5b-9 formed on GMC membrane could induce cell proliferation, the mechanism is still unclear. In this study, we first demonstrated that the level of SRY related HMG-BOX gene 9 (SOX9), general control nonderepressible 5 (GCN5), fibroblast growth factor 1 (FGF1) and platelet-derived growth factor α (PDGFα) was all elevated both in the renal tissues of Thy-1N rats (in vivo) and in the GMCs (in vitro) with sublytic C5b-9 stimulation. Then, we not only discovered that sublytic C5b-9 caused GMC proliferation through increasing SOX9, GCN5, FGF1 and PDGFα expression, but also proved that SOX9 and GCN5 formed a complex and combined with FGF1 and PDGFα promoters, leading to FGF1 and PDGFα gene transcription. More importantly, GCN5 could mediate SOX9 acetylation at lysine 62 (K62) to enhance SOX9 binding to FGF1 or PDGFα promoter and promote FGF1 or PDGFα synthesis and GMC proliferation. Besides, the experiments in vivo also showed that FGF1 and PDGFα expression, GMC proliferation and urinary protein secretion in Thy-1N rats were greatly reduced by silencing renal SOX9, GCN5, FGF1 or PDGFα gene. Furthermore, the renal tissues of MsPGN patients also exhibited positive expression of these genes mentioned above. Collectively, our findings indicate that GCN5, SOX9 and FGF1/PDGFα can form an axis and play an essential role in sublytic C5b-9-triggered GMC proliferation, which might provide a novel insight into the pathogenesis of Thy-1N and MsPGN.
Collapse
Affiliation(s)
- Longfei Liu
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China.,Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital, Nanjing Medical University, Huai'an, China
| | - Wen Ge
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Zhiwei Zhang
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Ya Li
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Mengxiao Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenhui Zhao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunlei Yao
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Can Luo
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Zhijiao Wu
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Wenbo Wang
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Dan Zhao
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Wen Qiu
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, China
| | - Yingwei Wang
- Key Laboratory of Immunological Environment and Disease, Department of Immunology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Li J, Chen X, Zhu L, Lao Z, Zhou T, Zang L, Ge W, Jiang M, Xu J, Cao Y, Du S, Yu Y, Fan G, Wang H. SOX9 is a critical regulator of TSPAN8-mediated metastasis in pancreatic cancer. Oncogene 2021; 40:4884-4893. [PMID: 34163029 PMCID: PMC8321899 DOI: 10.1038/s41388-021-01864-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/30/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the deadliest cancer mainly owing to its proclivity to early metastasis and the lack of effective targeted therapeutic drugs. Hence, understanding the molecular mechanisms underlying early invasion and metastasis by PDAC is imperative for improving patient outcomes. The present study identified that upregulation of TSPAN8 expression in PDAC facilitates metastasis in vivo and in vitro. We found SOX9 as a key transcriptional regulator of TSPAN8 expression in response to EGF stimulation. SOX9 modulation was sufficient to positively regulate endogenous expression of TSPAN8, with concomitant in vitro phenotypic changes such as loss of cell-matrix adherence and increased invasion. Moreover, increased SOX9 and TSPAN8 levels were shown to correlate in human pancreatic cancer specimens and downregulated in vitro by EGFR tyrosine kinase inhibitors. High expression of SOX9 and TSPAN8 has been associated with tumor stage, poor prognosis and poor patient survival in PDAC. In conclusion, this study highlights the importance of the EGF-SOX9-TSPAN8 signaling cascade in the control of PDAC invasion and implies that TSPAN8 may be a promising novel therapeutic target for the treatment of PDAC.
Collapse
Affiliation(s)
- Junjian Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoliang Chen
- The Center for Chronic Disease Control and Prevention, Shenzhen Guangming District Centers for Disease Control and Prevention, Shenzhen, China
| | - Liqun Zhu
- Department of Oncology, Liyang People's Hospital, Liyang, China
| | - Zhenghong Lao
- Department of Oncology, Deqing People's Hospital, Huzhou, China
| | - Tianhao Zhou
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijuan Zang
- Pathology Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiyu Ge
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyi Jiang
- Department of Medical Oncology, Shanghai Jiaotong University Affiliated Sixth People's Hospital East Campus, Shanghai, China
| | - Jingxuan Xu
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Cao
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoqian Du
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Yu
- Shanghai Experimental School, Shanghai, China
| | - Guangjian Fan
- Translational Medicine Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hongxia Wang
- State Key Laboratory of Oncogenes and Related Genes, Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Chen S, Yang L, Li Z, Zhuo S, Yan B, Zhang Z, Zhang J, Feng H, Yang K. EGFR/EGFRvIII partly regulates the tumourigenesis of glioblastoma through the SOX9-GLUT3 axis. Am J Transl Res 2021; 13:6055-6065. [PMID: 34306344 PMCID: PMC8290802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/29/2021] [Indexed: 06/13/2023]
Abstract
EGFR/EGFR variant III (EGFRvIII) glioblastoma is seriously malignant, and the underlying mechanism remains unclear. In this study, EGFR and GLUT3 were found to be co-expressed in our collected tissues and associated with worse overall survival in glioblastoma via bioinformatics analysis. Functionally, in vitro and in vivo tests revealed that silencing GLUT3 substantially inhibited the viability of U87-EGFRvIII and LN229-EGFRvIII cells. Compared with wild-type U87 or LN229 cells, the expression level of SOX9 in U87-EGFRvIII or LN229-EGFRvIII cells (U87 and LN229 over-expressing EGFRvIII) was substantially increased. Chromatin immunoprecipitation and Dual-luciferase reporter assays revealed that SOX9 bound to the promoter of GLUT3 and promoted the expression of GLUT3. Collectively, our findings indicated that the EGFR/EGFRvIII-SOX9-GLUT3 axis mediated the tumourigenesis of glioblastoma and might be a potential target for glioblastoma therapy.
Collapse
Affiliation(s)
- Shenbo Chen
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Liangwang Yang
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Zhengzheng Li
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Shenghua Zhuo
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Bo Yan
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Zhaoteng Zhang
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Jinben Zhang
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| | - Haizhong Feng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, China
| | - Kun Yang
- Department of Neurosurgery, The Affiliated Hospital of Hainan Medical CollegeHaikou 570102, Hainan, China
| |
Collapse
|
15
|
Chou WH, McGregor B, Schmidt A, Carvalho FLF, Hirsch MS, Chang SL, Kibel A, Mossanen M. Cyclophosphamide-associated bladder cancers and considerations for survivorship care: A systematic review. Urol Oncol 2021; 39:678-685. [PMID: 34134927 DOI: 10.1016/j.urolonc.2021.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/31/2021] [Accepted: 05/09/2021] [Indexed: 11/15/2022]
Abstract
PURPOSE We performed a systematic review to assess the clinical features of cyclophosphamide-associated bladder cancer. MATERIALS AND METHODS MEDLINE, Web of Science, and Cochrane Library were searched from inception to August 2020 according to PRISMA guidelines. Studies that associated bladder cancer with prior cyclophosphamide use on an individual level were included. RESULTS We identified 121 studies spanning over a 50-year period with 285 patients. The most common malignant indication for cyclophosphamide was lymphoma (25%), while the most common non-malignant indication was ANCA-associated vasculitides (26%). Hematuria and dysuria were the most prevalent symptoms prior to a cyclophosphamide-associated bladder cancer diagnosis, and median age at diagnosis was 55 years. Conventional urothelial carcinoma (UC) was the most common bladder-associated diagnosis (74%), although a broad range of cancer types were represented, notably leiomyosarcoma and squamous cell carcinoma. About half of bladder cancers were muscle invasive at diagnosis and median latency time was 10.0 years; 33% of patients had a bladder cancer related death. CONCLUSIONS We describe the largest pooled analysis of patients with cyclophosphamide-associated bladder cancer. These bladder cancers have a propensity for younger age at diagnosis, more advanced stage at diagnosis, and variant histology. There was a substantial number of patients with latency time of ≥20 years independent of cumulative cyclophosphamide dose. These findings support consideration of screening and long-term surveillance of cancer survivors with a history of cyclophosphamide therapy for bladder cancer.
Collapse
Affiliation(s)
- Wesley H Chou
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115
| | - Bradley McGregor
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215
| | - Andrew Schmidt
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215
| | - Filipe L F Carvalho
- Division of Urologic Surgery, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115
| | - Steven L Chang
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115; Division of Urologic Surgery, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115
| | - Adam Kibel
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215; Division of Urologic Surgery, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115
| | - Matthew Mossanen
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215; Division of Urologic Surgery, Brigham and Women's Hospital, 45 Francis Street, Boston, MA, 02115.
| |
Collapse
|
16
|
Identification of CRYAB + KCNN3 + SOX9 + Astrocyte-Like and EGFR + PDGFRA + OLIG1 + Oligodendrocyte-Like Tumoral Cells in Diffuse IDH1-Mutant Gliomas and Implication of NOTCH1 Signalling in Their Genesis. Cancers (Basel) 2021; 13:cancers13092107. [PMID: 33925547 PMCID: PMC8123787 DOI: 10.3390/cancers13092107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Diffuse grade II IDH-mutant gliomas are rare brain tumors mainly affecting young patients. These tumors are composed of different populations of tumoral cells. Little is known of these cells and how they are generated. These different cells may show different sensitivity to treatments, so our aim was to study them in detail by directly using patient resections. We identified two clearly distinct tumoral populations and defined reliable markers for them. We also uncovered part of the molecular mechanisms that generate them. Finally, we found that the two cell types have different electrical activity. This article provides unique data and new issues on these rare tumors, which need to be further investigated to develop innovative treatments. Abstract Diffuse grade II IDH-mutant gliomas are slow-growing brain tumors that progress into high-grade gliomas. They present intratumoral cell heterogeneity, and no reliable markers are available to distinguish the different cell subtypes. The molecular mechanisms underlying the formation of this cell diversity is also ill-defined. Here, we report that SOX9 and OLIG1 transcription factors, which specifically label astrocytes and oligodendrocytes in the normal brain, revealed the presence of two largely nonoverlapping tumoral populations in IDH1-mutant oligodendrogliomas and astrocytomas. Astrocyte-like SOX9+ cells additionally stained for APOE, CRYAB, ID4, KCNN3, while oligodendrocyte-like OLIG1+ cells stained for ASCL1, EGFR, IDH1, PDGFRA, PTPRZ1, SOX4, and SOX8. GPR17, an oligodendrocytic marker, was expressed by both cells. These two subpopulations appear to have distinct BMP, NOTCH1, and MAPK active pathways as stainings for BMP4, HEY1, HEY2, p-SMAD1/5 and p-ERK were higher in SOX9+ cells. We used primary cultures and a new cell line to explore the influence of NOTCH1 activation and BMP treatment on the IDH1-mutant glioma cell phenotype. This revealed that NOTCH1 globally reduced oligodendrocytic markers and IDH1 expression while upregulating APOE, CRYAB, HEY1/2, and an electrophysiologically-active Ca2+-activated apamin-sensitive K+ channel (KCNN3/SK3). This was accompanied by a reduction in proliferation. Similar effects of NOTCH1 activation were observed in nontumoral human oligodendrocytic cells, which additionally induced strong SOX9 expression. BMP treatment reduced OLIG1/2 expression and strongly upregulated CRYAB and NOGGIN, a negative regulator of BMP. The presence of astrocyte-like SOX9+ and oligodendrocyte-like OLIG1+ cells in grade II IDH1-mutant gliomas raises new questions about their role in the pathology.
Collapse
|
17
|
Akinduro OO, Suarez-Meade P, Garcia D, Brown DA, Sarabia-Estrada R, Attia S, Gokaslan ZL, Quiñones-Hinojosa A. Targeted Therapy for Chordoma: Key Molecular Signaling Pathways and the Role of Multimodal Therapy. Target Oncol 2021; 16:325-337. [PMID: 33893940 DOI: 10.1007/s11523-021-00814-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Chordoma is a rare but devastating tumor that arises in the cranial skull base or spine. There are currently no US Food and Drug Administration-approved targeted therapies for chordoma, and little understanding of whether using more than one therapy has benefit over monotherapy. OBJECTIVE The objective of this study was to systematically review the current status of clinical trials completed for patients with chordoma to determine if multimodal therapy offers a benefit in progression-free survival over monomodal therapy. METHODS We performed a systematic review of the literature according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines to review the available clinical trials of targeted therapy for chordoma. We compiled the clinical data to determine if there is a benefit of multimodal therapy over monotherapy. RESULTS Our search resulted in 11 clinical trials including 270 patients with advanced chordoma who were treated with targeted therapies. The most commonly employed targeted therapies acted within the following pathways: platelet-derived growth factor receptor (187 patients), vascular endothelial growth factor (66 patients), and mammalian target of rapamycin (43 patients). Reported progression-free survival for included studies ranged from 2.5 to 58 months, with the longest progression-free survival in a trial that included a platelet-derived growth factor receptor inhibitor, nilotinib, and concurrent radiotherapy (58.2 months). There was a higher range of progression-free survival for trials treating patients with multimodal therapy (10.2-14 months vs 2.5-9.2 months, except for a monotherapy trial published in 2020 with a progression-free survival of 18 months), and those published in 2018 or later (14-58.2 months vs 2.5-10.2 months). Only 23% of patients with chordoma in published clinical trials have been treated with multimodal therapy. CONCLUSIONS Progression-free survival may be enhanced by the use of targeted therapy with concurrent radiotherapy, use of multimodal therapy, and use of newer targeted therapy. Future clinical trials should consider use of concurrent radiotherapy and multimodal therapy for patients with advanced chordoma.
Collapse
Affiliation(s)
- Oluwaseun O Akinduro
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Paola Suarez-Meade
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Diogo Garcia
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | | | - Rachel Sarabia-Estrada
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA
| | - Steven Attia
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Ziya L Gokaslan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Alfredo Quiñones-Hinojosa
- Brain Tumor Stem Cell Laboratory, Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd. S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
18
|
Wang LB, Karpova A, Gritsenko MA, Kyle JE, Cao S, Li Y, Rykunov D, Colaprico A, Rothstein JH, Hong R, Stathias V, Cornwell M, Petralia F, Wu Y, Reva B, Krug K, Pugliese P, Kawaler E, Olsen LK, Liang WW, Song X, Dou Y, Wendl MC, Caravan W, Liu W, Cui Zhou D, Ji J, Tsai CF, Petyuk VA, Moon J, Ma W, Chu RK, Weitz KK, Moore RJ, Monroe ME, Zhao R, Yang X, Yoo S, Krek A, Demopoulos A, Zhu H, Wyczalkowski MA, McMichael JF, Henderson BL, Lindgren CM, Boekweg H, Lu S, Baral J, Yao L, Stratton KG, Bramer LM, Zink E, Couvillion SP, Bloodsworth KJ, Satpathy S, Sieh W, Boca SM, Schürer S, Chen F, Wiznerowicz M, Ketchum KA, Boja ES, Kinsinger CR, Robles AI, Hiltke T, Thiagarajan M, Nesvizhskii AI, Zhang B, Mani DR, Ceccarelli M, Chen XS, Cottingham SL, Li QK, Kim AH, Fenyö D, Ruggles KV, Rodriguez H, Mesri M, Payne SH, Resnick AC, Wang P, Smith RD, Iavarone A, Chheda MG, Barnholtz-Sloan JS, Rodland KD, Liu T, Ding L. Proteogenomic and metabolomic characterization of human glioblastoma. Cancer Cell 2021; 39:509-528.e20. [PMID: 33577785 PMCID: PMC8044053 DOI: 10.1016/j.ccell.2021.01.006] [Citation(s) in RCA: 326] [Impact Index Per Article: 108.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/02/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GBM) is the most aggressive nervous system cancer. Understanding its molecular pathogenesis is crucial to improving diagnosis and treatment. Integrated analysis of genomic, proteomic, post-translational modification and metabolomic data on 99 treatment-naive GBMs provides insights to GBM biology. We identify key phosphorylation events (e.g., phosphorylated PTPN11 and PLCG1) as potential switches mediating oncogenic pathway activation, as well as potential targets for EGFR-, TP53-, and RB1-altered tumors. Immune subtypes with distinct immune cell types are discovered using bulk omics methodologies, validated by snRNA-seq, and correlated with specific expression and histone acetylation patterns. Histone H2B acetylation in classical-like and immune-low GBM is driven largely by BRDs, CREBBP, and EP300. Integrated metabolomic and proteomic data identify specific lipid distributions across subtypes and distinct global metabolic changes in IDH-mutated tumors. This work highlights biological relationships that could contribute to stratification of GBM patients for more effective treatment.
Collapse
Affiliation(s)
- Liang-Bo Wang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Alla Karpova
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Dmitry Rykunov
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, USA; Division of Biostatistics, Department of Public Health Science, University of Miami, FL 33136, USA
| | - Joseph H Rothstein
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Vasileios Stathias
- Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, USA; Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; BD2K-LINCS Data Coordination and Integration Center, Miami, FL 33136, USA
| | - MacIntosh Cornwell
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yige Wu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karsten Krug
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Pietro Pugliese
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Emily Kawaler
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Lindsey K Olsen
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Wen-Wei Liang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Xiaoyu Song
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael C Wendl
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Mathematics, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Wenke Liu
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Daniel Cui Zhou
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jiayi Ji
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jamie Moon
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rosalie K Chu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Rui Zhao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Xiaolu Yang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | - Seungyeul Yoo
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexis Demopoulos
- Department of Neurology, Northwell Health System, Lake Success, NY 11042 USA
| | - Houxiang Zhu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Joshua F McMichael
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | | | - Caleb M Lindgren
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Hannah Boekweg
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Shuangjia Lu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jessika Baral
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Kelly G Stratton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Erika Zink
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Sneha P Couvillion
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Kent J Bloodsworth
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Weiva Sieh
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Simina M Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Stephan Schürer
- Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, USA; Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; BD2K-LINCS Data Coordination and Integration Center, Miami, FL 33136, USA; Institute for Data Science & Computing, University of Miami, FL 33136, USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63130, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Poznań University of Medical Sciences, 61-701 Poznań, Poland
| | | | - Emily S Boja
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Christopher R Kinsinger
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Tara Hiltke
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", 80128, Naples, Italy; BIOGEM, 83031 Ariano Irpino, Italy
| | - Xi S Chen
- Sylvester Comprehensive Cancer Center, University of Miami, FL 33136, USA; Division of Biostatistics, Department of Public Health Science, University of Miami, FL 33136, USA
| | - Sandra L Cottingham
- Department of Pathology, Spectrum Health and Helen DeVos Children's Hospital, Grand Rapids, MI 49503, USA
| | - Qing Kay Li
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Kelly V Ruggles
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Samuel H Payne
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Adam C Resnick
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA; Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Neurology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center and Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Research and Education, University Hospitals Health System, Cleveland, OH 44106, USA
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97221, USA.
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63130, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
19
|
Xie M, Wu Z, Ying S, Liu L, Zhao C, Yao C, Zhang Z, Luo C, Wang W, Zhao D, Zhang J, Qiu W, Wang Y. Sublytic C5b-9 induces glomerular mesangial cell proliferation via ERK1/2-dependent SOX9 phosphorylation and acetylation by enhancing Cyclin D1 in rat Thy-1 nephritis. Exp Mol Med 2021; 53:572-590. [PMID: 33811247 PMCID: PMC8102557 DOI: 10.1038/s12276-021-00589-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 02/01/2023] Open
Abstract
Glomerular mesangial cell (GMC) proliferation is a histopathological alteration in human mesangioproliferative glomerulonephritis (MsPGN) or in animal models of MsPGN, e.g., the rat Thy-1 nephritis (Thy-1N) model. Although sublytic C5b-9 assembly on the GMC membrane can trigger cell proliferation, the mechanisms are still undefined. We found that sublytic C5b-9-induced rat GMC proliferation was driven by extracellular signal-regulated kinase 1/2 (ERK1/2), sry-related HMG-box 9 (SOX9), and Cyclin D1. Here, ERK1/2 phosphorylation was a result of the calcium influx-PKC-α-Raf-MEK1/2 axis activated by sublytic C5b-9, and Cyclin D1 gene transcription was enhanced by ERK1/2-dependent SOX9 binding to the Cyclin D1 promoter (-582 to -238 nt). In addition, ERK1/2 not only interacted with SOX9 in the cell nucleus to mediate its phosphorylation at serine residues 64 (a new site identified by mass spectrometry) and 181 (a known site), but also indirectly induced SOX9 acetylation by elevating the expression of general control non-repressed protein 5 (GCN5), which together resulted in Cyclin D1 synthesis and GMC proliferation. Moreover, our in vivo experiments confirmed that silencing these genes ameliorated the lesions of Thy-1N rats and reduced SOX9 phosphorylation, acetylation and Cyclin D1 expression. Furthermore, the renal tissue sections of MsPGN patients also showed higher phosphorylation or expression of ERK1/2, SOX9, and Cyclin D1. In summary, these findings suggest that sublytic C5b-9-induced GMC proliferation in rat Thy-1N requires SOX9 phosphorylation and acetylation via enhanced Cyclin D1 gene transcription, which may provide a new insight into human MsPGN pathogenesis.
Collapse
Affiliation(s)
- Mengxiao Xie
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.412676.00000 0004 1799 0784Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Zhijiao Wu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Shuai Ying
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Longfei Liu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Department of Central Laboratory, The Affiliated Huaian No. 1 People’s Hospital, Nanjing Medical University, One West Huanghe Road, Huai’an, Jiangsu 223300 China
| | - Chenhui Zhao
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Chunlei Yao
- grid.412676.00000 0004 1799 0784Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029 China
| | - Zhiwei Zhang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Can Luo
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Wenbo Wang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Dan Zhao
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Jing Zhang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China
| | - Wen Qiu
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| | - Yingwei Wang
- grid.89957.3a0000 0000 9255 8984Department of Immunology, and Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166 China ,grid.89957.3a0000 0000 9255 8984Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166 China
| |
Collapse
|
20
|
So J, Kim M, Lee SH, Ko S, Lee DA, Park H, Azuma M, Parsons MJ, Prober D, Shin D. Attenuating the Epidermal Growth Factor Receptor-Extracellular Signal-Regulated Kinase-Sex-Determining Region Y-Box 9 Axis Promotes Liver Progenitor Cell-Mediated Liver Regeneration in Zebrafish. Hepatology 2021; 73:1494-1508. [PMID: 32602149 PMCID: PMC7769917 DOI: 10.1002/hep.31437] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS The liver is a highly regenerative organ, but its regenerative capacity is compromised in severe liver injury settings. In chronic liver diseases, the number of liver progenitor cells (LPCs) correlates proportionally to disease severity, implying that their inefficient differentiation into hepatocytes exacerbates the disease. Moreover, LPCs secrete proinflammatory cytokines; thus, their prolonged presence worsens inflammation and induces fibrosis. Promoting LPC-to-hepatocyte differentiation in patients with advanced liver disease, for whom liver transplantation is currently the only therapeutic option, may be a feasible clinical approach because such promotion generates more functional hepatocytes and concomitantly reduces inflammation and fibrosis. APPROACH AND RESULTS Here, using zebrafish models of LPC-mediated liver regeneration, we present a proof of principle of such therapeutics by demonstrating a role for the epidermal growth factor receptor (EGFR) signaling pathway in differentiation of LPCs into hepatocytes. We found that suppression of EGFR signaling promoted LPC-to-hepatocyte differentiation through the mitogen-activated ERK kinase (MEK)-extracellular signal-regulated kinase (ERK)-sex-determining region Y-box 9 (SOX9) cascade. Pharmacological inhibition of EGFR or MEK/ERK promoted LPC-to-hepatocyte differentiation as well as genetic suppression of the EGFR-ERK-SOX9 axis. Moreover, Sox9b overexpression in LPCs blocked their differentiation into hepatocytes. In the zebrafish liver injury model, both hepatocytes and biliary epithelial cells contributed to LPCs. EGFR inhibition promoted the differentiation of LPCs regardless of their origin. Notably, short-term treatment with EGFR inhibitors resulted in better liver recovery over the long term. CONCLUSIONS The EGFR-ERK-SOX9 axis suppresses LPC-to-hepatocyte differentiation during LPC-mediated liver regeneration. We suggest EGFR inhibitors as a proregenerative therapeutic drug for patients with advanced liver disease.
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Minwook Kim
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Seung-Hoon Lee
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sungjin Ko
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Present address: Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Daniel A. Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Hyewon Park
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Mizuki Azuma
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| | - Michael J. Parsons
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - David Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
21
|
Mirra S, Gavaldà-Navarro A, Manso Y, Higuera M, Serrat R, Salcedo MT, Burgaya F, Balibrea JM, Santamaría E, Uriarte I, Berasain C, Avila MA, Mínguez B, Soriano E, Villarroya F. ARMCX3 Mediates Susceptibility to Hepatic Tumorigenesis Promoted by Dietary Lipotoxicity. Cancers (Basel) 2021; 13:cancers13051110. [PMID: 33807672 PMCID: PMC7961652 DOI: 10.3390/cancers13051110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary An excess fat in the liver enhances the susceptibility to hepatic cancer. We found that Armcx3, a protein only known to date to play a role in neural development, is strongly increased in mouse liver in response to lipid availability and proliferation-inducing insults. In patients, the levels of hepatic Armcx3 are also increased in conditions of high exposure of the liver to fat. We wanted to determine the role of Armcx3 in the hepatocarcinogenesis favored by a high-fat diet. We generated mice with genetically driven suppression of Armcx3, and we found that they were protected against experimentally induced hepatic cancer, especially in conditions of a high-fat diet. Armcx3 was also found to promote hepatic cell proliferation through the interaction with Sox9, a known proliferation factor in hepatocellular carcinoma. Armcx3 is identified as a novel factor in meditating propensity to liver cancer in conditions of high hepatic lipid insults. Abstract ARMCX3 is encoded by a member of the Armcx gene family and is known to be involved in nervous system development and function. We found that ARMCX3 is markedly upregulated in mouse liver in response to high lipid availability, and that hepatic ARMCX3 is upregulated in patients with NAFLD and hepatocellular carcinoma (HCC). Mice were subjected to ARMCX3 invalidation (inducible ARMCX3 knockout) and then exposed to a high-fat diet and diethylnitrosamine-induced hepatocarcinogenesis. The effects of experimental ARMCX3 knockdown or overexpression in HCC cell lines were also analyzed. ARMCX3 invalidation protected mice against high-fat-diet-induced NAFLD and chemically induced hepatocarcinogenesis. ARMCX3 invalidation promoted apoptotic cell death and macrophage infiltration in livers of diethylnitrosamine-treated mice maintained on a high-fat diet. ARMCX3 downregulation reduced the viability, clonality and migration of HCC cell lines, whereas ARMCX3 overexpression caused the reciprocal effects. SOX9 was found to mediate the effects of ARMCX3 in hepatic cells, with the SOX9 interaction required for the effects of ARMCX3 on hepatic cell proliferation. In conclusion, ARMCX3 is identified as a novel molecular actor in liver physiopathology and carcinogenesis. ARMCX3 downregulation appears to protect against hepatocarcinogenesis, especially under conditions of high dietary lipid-mediated hepatic insult.
Collapse
Affiliation(s)
- Serena Mirra
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08007 Barcelona, Spain; (S.M.); (Y.M.); (R.S.); (F.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Aleix Gavaldà-Navarro
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Yasmina Manso
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08007 Barcelona, Spain; (S.M.); (Y.M.); (R.S.); (F.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mónica Higuera
- Liver Diseases Research Group, Vall d’Hebron Institute of Research, VHIR, 08035 Barcelona, Spain; (M.H.); (B.M.)
| | - Román Serrat
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08007 Barcelona, Spain; (S.M.); (Y.M.); (R.S.); (F.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Teresa Salcedo
- Pathology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Ferran Burgaya
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08007 Barcelona, Spain; (S.M.); (Y.M.); (R.S.); (F.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Maria Balibrea
- Endocrine, Metabolic and Bariatric Surgery Unit, General Surgery Department, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain;
| | - Eva Santamaría
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.S.);(I.U.); (C.B.); (M.A.A.)
- Hepatology Programme, CIMA-University of Navarra, IdiSNA, 31009 Pamplona, Spain
| | - Iker Uriarte
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.S.);(I.U.); (C.B.); (M.A.A.)
- Hepatology Programme, CIMA-University of Navarra, IdiSNA, 31009 Pamplona, Spain
| | - Carmen Berasain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.S.);(I.U.); (C.B.); (M.A.A.)
- Hepatology Programme, CIMA-University of Navarra, IdiSNA, 31009 Pamplona, Spain
| | - Matias A. Avila
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.S.);(I.U.); (C.B.); (M.A.A.)
- Hepatology Programme, CIMA-University of Navarra, IdiSNA, 31009 Pamplona, Spain
| | - Beatriz Mínguez
- Liver Diseases Research Group, Vall d’Hebron Institute of Research, VHIR, 08035 Barcelona, Spain; (M.H.); (B.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain; (E.S.);(I.U.); (C.B.); (M.A.A.)
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08007 Barcelona, Spain; (S.M.); (Y.M.); (R.S.); (F.B.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (E.S.); (F.V.); Tel.: +34-934-037-117 (E.S.); +34-934-021-525 (F.V.)
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (E.S.); (F.V.); Tel.: +34-934-037-117 (E.S.); +34-934-021-525 (F.V.)
| |
Collapse
|
22
|
Panda M, Tripathi SK, Biswal BK. SOX9: An emerging driving factor from cancer progression to drug resistance. Biochim Biophys Acta Rev Cancer 2021; 1875:188517. [PMID: 33524528 DOI: 10.1016/j.bbcan.2021.188517] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of transcription factors is one of the common problems in the pathogenesis of human cancer. Among them, SOX9 is one of the critical transcription factors involved in various diseases, including cancer. The expression of SOX9 is regulated by microRNAs (miRNAs), methylation, phosphorylation, and acetylation. Interestingly, SOX9 acts as a proto-oncogene or tumor suppressor gene, relying upon kinds of cancer. Recent studies have reported the critical role of SOX9 in the regulation of the tumor microenvironment (TME). Additionally, activation of SOX9 signaling or SOX9 regulated signaling pathways play a crucial role in cancer development and progression. Accumulating evidence also suggests that SOX9 acquires stem cell features to induce epithelial-mesenchymal transition (EMT). Moreover, SOX9 has been broadly studied in the field of cancer stem cell (CSC) and EMT in the last decades. However, the link between SOX9 and cancer drug resistance has only recently been discovered. Furthermore, its differential expression could be a potential biomarker for tumor prognosis and progression. This review outlined the various biological implications of SOX9 in cancer progression and cancer drug resistance and elucidated its signaling network, which could be a potential target for designing novel anticancer drugs.
Collapse
Affiliation(s)
- Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| |
Collapse
|
23
|
Kaushik G, Seshacharyulu P, Rauth S, Nallasamy P, Rachagani S, Nimmakayala RK, Vengoji R, Mallya K, Chirravuri-Venkata R, Singh AB, Foster JM, Ly QP, Smith LM, Lele SM, Malafa MP, Jain M, Ponnusamy MP, Batra SK. Selective inhibition of stemness through EGFR/FOXA2/SOX9 axis reduces pancreatic cancer metastasis. Oncogene 2020; 40:848-862. [PMID: 33288882 PMCID: PMC7848971 DOI: 10.1038/s41388-020-01564-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer (PC) is difficult to defeat due to mechanism (s) driving metastasis and drug resistance. Cancer stemness is a major challenging phenomenon associated with PC metastasis and limiting therapy efficacy. In this study, we evaluated the pre-clinical and clinical significance of eradicating pancreatic cancer stem cells (PCSC) and its components using a pan-EGFR inhibitor afatinib in combination with gemcitabine. Afatinib in combination with gemcitabine, significantly reduced KrasG12D/+; Pdx-1 Cre (KC) (P<0.01) and KrasG12D/+; p53R172H/+; Pdx-1 Cre (KPC) (P<0.05) derived mouse tumoroids and KPC-derived murine syngeneic cell line growth compared to gemcitabine/afatinib alone treatment. The drug combination also reduced PC xenograft tumor burden (P<0.05) and the incidence of metastasis by affecting key stemness markers, as confirmed by co-localization studies. Moreover, the drug combination significantly decreases the growth of various PC patient-derived organoids (P<0.001). We found that SOX9 is significantly overexpressed in high-grade PC tumors (P<0.05) and in chemotherapy-treated patients compared to chemo-naïve patients (P<0.05). These results were further validated using publicly available datasets. Moreover, afatinib alone or in combination with gemcitabine decreased stemness and tumorspheres by reducing phosphorylation of EGFR family proteins, ERK, FAK, and CSC markers. Mechanistically, afatinib treatment decreased CSC markers by downregulating SOX9 via FOXA2. Indeed, EGFR and FOXA2 depletion reduced SOX9 expression in PCSCs. Taken together, pan EGFR inhibition by afatinib impedes PCSCs growth and metastasis via the EGFR/ERK/FOXA2/SOX9 axis. This novel mechanism of panEGFR inhibitor and its ability to eradicate CSC may serve as a tailor-made approach to enhance chemotherapeutic benefits in other cancer types.
Collapse
Affiliation(s)
- Garima Kaushik
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jason M Foster
- Division of Surgical Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Quan P Ly
- Division of Surgical Oncology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Subodh M Lele
- Department of Pathology and Microbiology, UNMC, Omaha, NE, USA
| | - Mokenge P Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pathology and Microbiology, UNMC, Omaha, NE, USA. .,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
24
|
Rose M, Maurer A, Wirtz J, Bleilevens A, Waldmann T, Wenz M, Eyll M, Geelvink M, Gereitzig M, Rüchel N, Denecke B, Eltze E, Herrmann E, Toma M, Horst D, Grimm T, Denzinger S, Ecke T, Vögeli TA, Knuechel R, Maurer J, Gaisa NT. EGFR activity addiction facilitates anti-ERBB based combination treatment of squamous bladder cancer. Oncogene 2020; 39:6856-6870. [PMID: 32978523 PMCID: PMC7605436 DOI: 10.1038/s41388-020-01465-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/11/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022]
Abstract
Recent findings suggested a benefit of anti-EGFR therapy for basal-like muscle-invasive bladder cancer (MIBC). However, the impact on bladder cancer with substantial squamous differentiation (Sq-BLCA) and especially pure squamous cell carcinoma (SCC) remains unknown. Therefore, we comprehensively characterized pure and mixed Sq-BLCA (n = 125) on genetic and protein expression level, and performed functional pathway and drug-response analyses with cell line models and isolated primary SCC (p-SCC) cells of the human urinary bladder. We identified abundant EGFR expression in 95% of Sq-BLCA without evidence for activating EGFR mutations. Both SCaBER and p-SCC cells were sensitive to EGFR tyrosine kinase inhibitors (TKIs: erlotinib and gefitinib). Combined treatment with anti-EGFR TKIs and varying chemotherapeutics led to a concentration-dependent synergism in SCC cells according to the Chou-Talalay method. In addition, the siRNA knockdown of EGFR impaired SCaBER viability suggesting a putative "Achilles heel" of Sq-BLCA. The observed effects seem Sq-BLCA-specific since non-basal urothelial cancer cells were characterized by poor TKI sensitivity associated with a short-term feedback response potentially attenuating anti-tumor activity. Hence, our findings give further insights into a crucial, Sq-BLCA-specific role of the ERBB signaling pathway proposing improved effectiveness of anti-EGFR based regimens in combination with chemotherapeutics in squamous bladder cancers with wild-type EGFR-overexpression.
Collapse
MESH Headings
- Aged
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/genetics
- Carcinoma, Transitional Cell/pathology
- Cell Line, Tumor
- Cohort Studies
- Drug Resistance, Neoplasm/drug effects
- Drug Synergism
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Erlotinib Hydrochloride/pharmacology
- Erlotinib Hydrochloride/therapeutic use
- Female
- Gefitinib/pharmacology
- Gefitinib/therapeutic use
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Male
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- RNA, Small Interfering/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-4/antagonists & inhibitors
- Receptor, ErbB-4/metabolism
- Signal Transduction/drug effects
- Urinary Bladder/pathology
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Angela Maurer
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Julia Wirtz
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | | | - Tanja Waldmann
- Department of Gynecology, University Clinic RWTH, Aachen, Germany
| | - Maximilian Wenz
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Marie Eyll
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Mirja Geelvink
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | | | - Nadine Rüchel
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- IZKF Aachen, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Elke Eltze
- Institute of Pathology, Saarbrücken-Rastpfuhl, Saarbrücken, Germany
| | - Edwin Herrmann
- Department of Urology, University Hospital Münster, Münster, Germany
| | - Marieta Toma
- Institute of Pathology, University Hospital Gustav Carus TU Dresden, Dresden, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Grimm
- Department of Urology, LMU Munich, Munich, Germany
| | - Stefan Denzinger
- Department of Urology, University of Regensburg, Regensburg, Germany
| | - Thorsten Ecke
- Department of Urology, Helios Hospital Bad Saarow, Bad Saarow, Germany
| | | | - Ruth Knuechel
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Jochen Maurer
- Department of Gynecology, University Clinic RWTH, Aachen, Germany
| | - Nadine T Gaisa
- Institute of Pathology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
25
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
26
|
Ma Y, Shepherd J, Zhao D, Bollu LR, Tahaney WM, Hill J, Zhang Y, Mazumdar A, Brown PH. SOX9 Is Essential for Triple-Negative Breast Cancer Cell Survival and Metastasis. Mol Cancer Res 2020; 18:1825-1838. [PMID: 32661114 DOI: 10.1158/1541-7786.mcr-19-0311] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/21/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
Triple-negative breast cancer (TNBC) has the worst prognosis of all breast cancers, and lacks effective targeted treatment strategies. Previously, we identified 33 transcription factors highly expressed in TNBC. Here, we focused on six sex determining region Y-related HMG-box (SOX) transcription factors (SOX4, 6, 8, 9, 10, and 11) highly expressed in TNBCs. Our siRNA screening assay demonstrated that SOX9 knockdown suppressed TNBC cell growth and invasion in vitro. Thus, we hypothesized that SOX9 is an important regulator of breast cancer survival and metastasis, and demonstrated that knockout of SOX9 reduced breast tumor growth and lung metastasis in vivo. In addition, we found that loss of SOX9 induced profound apoptosis, with only a slight impairment of G1 to S progression within the cell cycle, and that SOX9 directly regulates genes controlling apoptosis. On the basis of published CHIP-seq data, we demonstrated that SOX9 binds to the promoter of apoptosis-regulating genes (tnfrsf1b, fadd, tnfrsf10a, tnfrsf10b, and ripk1), and represses their expression. SOX9 knockdown upregulates these genes, consistent with the induction of apoptosis. Analysis of available CHIP-seq data showed that SOX9 binds to the promoters of several epithelial-mesenchymal transition (EMT)- and metastasis-regulating genes. Using CHIP assays, we demonstrated that SOX9 directly binds the promoters of genes involved in EMT (vim, cldn1, ctnnb1, and zeb1) and that SOX9 knockdown suppresses the expression of these genes. IMPLICATIONS: Our studies identified the SOX9 protein as a "master regulator" of breast cancer cell survival and metastasis, and provide preclinical rationale to develop SOX9 inhibitors for the treatment of women with metastatic triple-negative breast cancer.
Collapse
Affiliation(s)
- Yanxia Ma
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Jonathan Shepherd
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Dekuang Zhao
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Lakshmi Reddy Bollu
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - William M Tahaney
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Yun Zhang
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas M.D. Anderson Cancer Center, Houston, Texas. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.,Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
27
|
Pastore N, Huynh T, Herz NJ, Calcagni' A, Klisch TJ, Brunetti L, Kim KH, De Giorgi M, Hurley A, Carissimo A, Mutarelli M, Aleksieva N, D'Orsi L, Lagor WR, Moore DD, Settembre C, Finegold MJ, Forbes SJ, Ballabio A. TFEB regulates murine liver cell fate during development and regeneration. Nat Commun 2020; 11:2461. [PMID: 32424153 PMCID: PMC7235048 DOI: 10.1038/s41467-020-16300-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
It is well established that pluripotent stem cells in fetal and postnatal liver (LPCs) can differentiate into both hepatocytes and cholangiocytes. However, the signaling pathways implicated in the differentiation of LPCs are still incompletely understood. Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, is known to be involved in osteoblast and myeloid differentiation, but its role in lineage commitment in the liver has not been investigated. Here we show that during development and upon regeneration TFEB drives the differentiation status of murine LPCs into the progenitor/cholangiocyte lineage while inhibiting hepatocyte differentiation. Genetic interaction studies show that Sox9, a marker of precursor and biliary cells, is a direct transcriptional target of TFEB and a primary mediator of its effects on liver cell fate. In summary, our findings identify an unexplored pathway that controls liver cell lineage commitment and whose dysregulation may play a role in biliary cancer.
Collapse
Affiliation(s)
- Nunzia Pastore
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Tuong Huynh
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Niculin J Herz
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alessia Calcagni'
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tiemo J Klisch
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lorenzo Brunetti
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kangho Ho Kim
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Marco De Giorgi
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayrea Hurley
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Annamaria Carissimo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
| | | | - Niya Aleksieva
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Luca D'Orsi
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
| | - William R Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David D Moore
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, 80131, Italy
| | - Milton J Finegold
- Department of Pathology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Andrea Ballabio
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, NA, 80078, Italy.
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, 80131, Italy.
| |
Collapse
|
28
|
Ray B, Maloney B, Sambamurti K, Karnati HK, Nelson PT, Greig NH, Lahiri DK. Rivastigmine modifies the α-secretase pathway and potentially early Alzheimer's disease. Transl Psychiatry 2020; 10:47. [PMID: 32066688 PMCID: PMC7026402 DOI: 10.1038/s41398-020-0709-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Rivastigmine (or Exelon) is a cholinesterase inhibitor, currently used as a symptomatic treatment for mild-to-moderate Alzheimer's disease (AD). Amyloid-β peptide (Aβ) generated from its precursor protein (APP) by β-secretase (or BACE1) and γ-secretase endoproteolysis. Alternative APP cleavage by α-secretase (a family of membrane-bound metalloproteases- Adamalysins) precludes the generation of toxic Aβ and yields a neuroprotective and neurotrophic secreted sAPPα fragment. Several signal transduction pathways, including protein kinase C and MAP kinase, stimulate α-secretase. We present data to suggest that rivastigmine, in addition to anticholinesterase activity, directs APP processing away from BACE1 and towards α-secretases. We treated rat neuronal PC12 cells and primary human brain (PHB) cultures with rivastigmine and the α-secretase inhibitor TAPI and assayed for levels of APP processing products and α-secretases. We subsequently treated 3×Tg (transgenic) mice with rivastigmine and harvested hippocampi to assay for levels of APP processing products. We also assayed postmortem human control, AD, and AD brains from subjects treated with rivastigmine for levels of APP metabolites. Rivastigmine dose-dependently promoted α-secretase activity by upregulating levels of ADAM-9, -10, and -17 α-secretases in PHB cultures. Co-treatment with TAPI eliminated rivastigmine-induced sAPPα elevation. Rivastigmine treatment elevated levels of sAPPα in 3×Tg mice. Consistent with these results, we also found elevated sAPPα in postmortem brain samples from AD patients treated with rivastigmine. Rivastigmine can modify the levels of several shedding proteins and directs APP processing toward the non-amyloidogenic pathway. This novel property of rivastigmine can be therapeutically exploited for disease-modifying intervention that goes beyond symptomatic treatment for AD.
Collapse
Affiliation(s)
- Balmiki Ray
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Bryan Maloney
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Kumar Sambamurti
- grid.259828.c0000 0001 2189 3475Department of Neurosciences, Medical University of South Carolina, Charleston, 29425 SC USA
| | - Hanuma K. Karnati
- grid.419475.a0000 0000 9372 4913National Institute on Aging, Drug Design and Development Section, Bethesda, MD 20892 USA
| | - Peter T. Nelson
- grid.266539.d0000 0004 1936 8438Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536 USA
| | - Nigel H. Greig
- grid.419475.a0000 0000 9372 4913National Institute on Aging, Drug Design and Development Section, Bethesda, MD 20892 USA
| | - Debomoy K. Lahiri
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
29
|
Schneider AJ, Gawdzik J, Vezina CM, Baker TR, Peterson RE. Sox9 in mouse urogenital sinus epithelium mediates elongation of prostatic buds and expression of genes involved in epithelial cell migration. Gene Expr Patterns 2019; 34:119075. [PMID: 31669249 PMCID: PMC6927329 DOI: 10.1016/j.gep.2019.119075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/23/2022]
Abstract
Previous studies identified Sox9 as a critical mediator of prostate development but the precise stage when Sox9 acts had not been determined. A genetic approach was used to delete Sox9 from mouse urogenital sinus epithelium (UGE) prior to prostate specification. All prostatic bud types (anterior, dorsolateral and ventral) were stunted in Sox9 conditional knockouts (cKOs) even though the number of prostatic buds did not differ from that of controls. We concluded that Sox9 is required for prostatic bud elongation and compared control male, control female, Sox9 cKO male and Sox9 cKO female UGE transcriptomes to identify potential molecular mediators. We identified 702 sex-dependent and 95 Sox9-dependent genes. Thirty-one genes were expressed in both a sex- and Sox9-dependent pattern. A comparison of Sox9 cKO female vs control female UGE transcriptomes revealed 74 Sox9-dependent genes, some of which also function in cell migration. SOX9 regulates, directly or indirectly, a largely different profile of genes in male and female UGE. Eighty-three percent of Sox9-dependent genes in male UGE were not Sox9-dependent in female UGE. Only 16 genes were Sox9-dependent in the UGE of both sexes and seven had cell migration functions. These results support the notion that Sox9 promotes cell migration activities needed for prostate ductal elongation.
Collapse
Affiliation(s)
- Andrew J Schneider
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA.
| | - Joseph Gawdzik
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA; Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI, 53706, USA.
| | - Chad M Vezina
- School of Veterinary Medicine, University of Wisconsin-Madison, 1656 Linden Drive, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI, 53706, USA.
| | - Tracie R Baker
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI, 53706, USA; Institute of Environmental Health Sciences and School of Medicine, Wayne State University, 6135 Woodward Avenue, Detroit, MI, 48202, USA.
| | - Richard E Peterson
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI, 53705, USA; Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, 1400 University Avenue, Madison, WI, 53706, USA.
| |
Collapse
|
30
|
Zhang W, Wu Y, Hou B, Wang Y, Deng D, Fu Z, Xu Z. A SOX9-AS1/miR-5590-3p/SOX9 positive feedback loop drives tumor growth and metastasis in hepatocellular carcinoma through the Wnt/β-catenin pathway. Mol Oncol 2019; 13:2194-2210. [PMID: 31402556 PMCID: PMC6763786 DOI: 10.1002/1878-0261.12560] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/18/2019] [Accepted: 08/09/2019] [Indexed: 01/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent solid tumor with a high global death rate. SRY box 9 (SOX9) has been reported as an oncogene in HCC by several studies, but the underlying mechanism remains largely unexplored. Here, we confirmed upregulation of SOX9 in HCC tissues and cell lines and validated that SOX9 facilitates proliferation, migration and invasion in HCC. We subsequently identified that the long non‐coding RNA (lncRNA) SOX9 antisense RNA 1 (SOX9‐AS1) is a neighbor gene to SOX9; SOX9‐AS1 is also upregulated in HCC, and its expression is positively correlated with that of SOX9. In addition, SOX9‐AS1 appears to have prognostic significance in HCC patients. We showed that SOX9‐AS1 aggravates HCC progression and metastasis in vitro and in vivo. We demonstrated that SOX9‐AS1 sponges miR‐5590‐3p to elevate SOX9 expression, and that SOX9 in turn transcriptionally activates SOX9‐AS1. Moreover, we verified that SOX9‐AS1 regulates SOX9 and its known downstream Wnt/β‐catenin pathway so as to facilitate epithelial‐to‐mesenchymal transition. The results of our rescue assays suggest that SOX9‐AS1 regulates HCC progression through SOX9 and the Wnt/β‐catenin pathway. In conclusion, our study demonstrates that a SOX9‐AS1/miR‐5590‐3p/SOX9 positive feedback loop drives tumor growth and metastasis in HCC through the Wnt/β‐catenin pathway, suggesting SOX9‐AS1 as a novel potential prognostic and treatment target for HCC.
Collapse
Affiliation(s)
- Wei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanhui Wu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bingwu Hou
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yadong Wang
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| | - Dongfeng Deng
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| | - Zhihao Fu
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| | - Zongquan Xu
- Department of Hepatobiliary Pancreatic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan, China
| |
Collapse
|
31
|
Kumar P, Mistri TK. Transcription factors in SOX family: Potent regulators for cancer initiation and development in the human body. Semin Cancer Biol 2019; 67:105-113. [PMID: 31288067 DOI: 10.1016/j.semcancer.2019.06.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Transcription factors (TFs) have a key role in controlling the gene regulatory network that sustains explicit cell states in humans. However, an uncontrolled regulation of these genes potentially results in a wide range of diseases, including cancer. Genes of the SOX family are indeed crucial as deregulation of SOX family TFs can potentially lead to changes in cell fate as well as irregular cell growth. SOX TFs are a conserved group of transcriptional regulators that mediate DNA binding through a highly conserved high-mobility group (HMG) domain. Accumulating evidence demonstrates that cell fate and differentiation in major developmental processes are controlled by SOX TFs. Besides; numerous reports indicate that both up- and down-regulation of SOX TFs may induce cancer progression. In this review, we discuss the involvement of key TFs of SOX family in human cancers.
Collapse
Affiliation(s)
- Prasann Kumar
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Agronomy, Lovely Professional University, Jalandhar, Punjab, 144411, India
| | - Tapan Kumar Mistri
- The Division of Research and Development, Lovely Professional University, Jalandhar, Punjab, 144411, India; The Department of Chemistry, Lovely Professional University, Jalandhar, Punjab, 144411, India.
| |
Collapse
|
32
|
da Cruz RS, Clarke J, Curi ACP, Al-Yawar A, Jin L, Baird A, Cruz MI, Kallakury B, de Assis S. Parental obesity programs pancreatic cancer development in offspring. Endocr Relat Cancer 2019; 26:511-523. [PMID: 30865925 PMCID: PMC6717698 DOI: 10.1530/erc-19-0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Epidemiological studies suggest that timing of obesity onset - and underlying metabolic dysfunction - is important in determining pancreatic cancer rates: early and young adult abdominal overweight/obesity is more strongly associated with this cancer than obesity that develops later in life. Parental obesity and overweight are associated with metabolic dysfunction and obesity in their children. Here, we evaluated the impact of parental overweight on offspring's susceptibility of pancreatic cancer using the P48Cre/+/KrasG12D/+ mouse model. Male mice were fed an obesity-inducing diet (OID) before conception and mated with females raised on a control diet (CO) to generate the offspring. In a separate experiment, pregnant dams were fed CO or OID throughout gestation. The resulting OID offspring from the maternal (OID-m) or paternal lineage (OID-p) were used to study body weight, metabolic parameters and pancreatic cancer development and for molecular analysis. Parental obesity increased offspring's body weight at birth, weaning and in adulthood compared to CO, with gender- and genotype-specific differences. OID-p and OID-m offspring showed metabolic disorder and accelerated development of high-grade PanIN/PDAC. OID offspring also had higher rates of acinar-to-ductal reprogramming assessed by CPA1+/SOX9+-positive pancreatic cells. Levels of Tenascin C (TNC), an ECM glycoprotein shown to suppress apoptosis, were elevated in OID offspring, particularly females. In line with that, OID offspring displayed increased collagen content and decreased apoptosis in pancreatic lesions compared to CO. An ancestral history of obesity through either the paternal or maternal lineages increases offspring's susceptibility to pancreatic cancer development.
Collapse
|
33
|
Domenici G, Aurrekoetxea-Rodríguez I, Simões BM, Rábano M, Lee SY, Millán JS, Comaills V, Oliemuller E, López-Ruiz JA, Zabalza I, Howard BA, Kypta RM, Vivanco MDM. A Sox2-Sox9 signalling axis maintains human breast luminal progenitor and breast cancer stem cells. Oncogene 2019; 38:3151-3169. [PMID: 30622340 PMCID: PMC6756022 DOI: 10.1038/s41388-018-0656-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/12/2018] [Accepted: 12/07/2018] [Indexed: 01/02/2023]
Abstract
Increased cancer stem cell content during development of resistance to tamoxifen in breast cancer is driven by multiple signals, including Sox2-dependent activation of Wnt signalling. Here, we show that Sox2 increases and estrogen reduces the expression of the transcription factor Sox9. Gain and loss of function assays indicate that Sox9 is implicated in the maintenance of human breast luminal progenitor cells. CRISPR/Cas knockout of Sox9 reduces growth of tamoxifen-resistant breast tumours in vivo. Mechanistically, Sox9 acts downstream of Sox2 to control luminal progenitor cell content and is required for expression of the cancer stem cell marker ALDH1A3 and Wnt signalling activity. Sox9 is elevated in breast cancer patients after endocrine therapy failure. This new regulatory axis highlights the relevance of SOX family transcription factors as potential therapeutic targets in breast cancer.
Collapse
Affiliation(s)
| | | | - Bruno M Simões
- CIC bioGUNE, Technological Park Bizkaia, Derio, 48160, Spain
| | - Miriam Rábano
- CIC bioGUNE, Technological Park Bizkaia, Derio, 48160, Spain
| | - So Young Lee
- CIC bioGUNE, Technological Park Bizkaia, Derio, 48160, Spain
| | | | | | - Erik Oliemuller
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Ignacio Zabalza
- Department of Pathology, Galdakao-Usansolo Hospital, Galdakao, Spain
| | - Beatrice A Howard
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Robert M Kypta
- CIC bioGUNE, Technological Park Bizkaia, Derio, 48160, Spain.,Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
34
|
Abstract
Urothelial carcinoma is a tumor type featuring pronounced intertumoral heterogeneity and a high mutational and epigenetic load. The two major histopathological urothelial carcinoma types - the non-muscle-invasive and muscle-invasive urothelial carcinoma - markedly differ in terms of their respective typical mutational profiles and also by their probable cells of origin, that is, a urothelial basal cell for muscle-invasive carcinomas and a urothelial intermediate cell for at least a large part of non-muscle-invasive carcinomas. Both non-muscle-invasive and muscle-invasive urothelial carcinomas can be further classified into discrete intrinsic subtypes based on their typical transcriptomic profiles. Urothelial carcinogenesis shows a number of parallels to a urothelial regenerative response. Both of these processes seem to be dominated by specific stem cell populations. In the last years, the nature and location of urothelial stem cell(s) have been subject to many controversies, which now seem to be settled down, favoring the existence of a largely single urothelial stem cell type located among basal cells. Basal cell markers have also been amply used to identify urothelial carcinoma stem cells, especially in muscle-invasive disease, but they proved useful even in some non-muscle-invasive tumors. Analyses on molecular nature of urothelial carcinoma stem cells performed till now point to their great heterogeneity, both during the tumor development and upon intertumoral comparison, sexual dimorphism providing a special example of the latter. Moreover, urothelial cancer stem cells are endowed with intrinsic plasticity, whereby they can modulate their stemness in relation to other tumor-related traits, especially motility and invasiveness. Such transitional modulations suggest underlying epigenetic mechanisms and, even within this context, inter- and intratumoral heterogeneity becomes apparent. Multiple molecular aspects of urothelial cancer stem cell biology markedly influence therapeutic response, implying their knowledge as a prerequisite to improved therapies of this disease. At the same time, the notion of urothelial cancer stem cell heterogeneity implies that this therapeutic benefit would be most probably and most efficiently achieved within the context of individualized antitumor therapy.
Collapse
|
35
|
Janssen JN, Batschkus S, Schimmel S, Bode C, Schminke B, Miosge N. The Influence of TGF-β3, EGF, and BGN on SOX9 and RUNX2 Expression in Human Chondrogenic Progenitor Cells. J Histochem Cytochem 2018; 67:117-127. [PMID: 30431382 DOI: 10.1369/0022155418811645] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease and leads to the degradation of the extracellular matrix by an imbalance between anabolic and catabolic processes. TGF-β3 (transforming growth factor beta-3) and epidermal growth factor (EGF) influence the osteochondrogenic potential of chondrocytes. In this study, we compared the expression of mediators and receptors in the TGF-β3 and EGF pathways, as well as biglycan (BGN), in healthy and diseased chondrocytes. Furthermore, we used chondrogenic progenitor cells (CPCs) for in vitro stimulation and knockdown experiments to elucidate the effects of TGF-β3 and EGF on the chondrogenic potential. Our results demonstrate that the expression of TGF-beta receptor type-1 (TGFBRI) and epidermal growth factor receptor (EGFR) is altered in diseased chondrocytes as well as in CPCs. Moreover, TGF-β3 and EGF stimulation influenced the expression levels of BGN, SRY (sex determining region Y)-box 9 (SOX9), and Runt-related transcription factor 2 (RUNX2) in CPCs. Therefore, changes in TGFBRI and EGFR expression likely contribute to the degenerative and regenerative effects seen in late stages of OA.
Collapse
Affiliation(s)
| | | | - Stefan Schimmel
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Christa Bode
- Tissue Regeneration Work Group, Department of Prosthodontics
| | - Boris Schminke
- Department of Oral and Maxillofacial Surgery, University Medical Center, Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics
| |
Collapse
|
36
|
Lin S, Liu K, Zhang Y, Jiang M, Lu R, Folts CJ, Gao X, Noble MD, Zhao T, Zhou Z, Lan X, Que J. Pharmacological targeting of p38 MAP-Kinase 6 (MAP2K6) inhibits the growth of esophageal adenocarcinoma. Cell Signal 2018; 51:222-232. [PMID: 30102978 DOI: 10.1016/j.cellsig.2018.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 07/20/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
Drug repurposing with a better understanding of the underlying mechanism has provided new avenues to find treatment for malignancies. Esophageal adenocarcinoma (EAC) is a rapidly increasing cancer with a dismal 5-year survival rate of <15%. Lack of efficient treatment options contributes to the high mortality rate of EAC. To find new therapy against EAC we performed unbiased drug screening of an FDA-approved drug library and identified that the cardiac glycosides including Ouabain, Digoxin and Digitoxin efficiently inhibit the proliferation of EAC cell lines (OE33 and OE19) both in vitro and in vivo. RNA-Sequencing analysis combined with RNAi screening revealed that Ouabain suppresses the proliferation of EAC cells through downregulation of p38 MAP-Kinase 6 (MAP2K6, also known as MKK6). Consistently, shRNA-mediated knockdown of MKK6 reduced the proliferation of EAC cells and tumor growth. Further analysis demonstrated that MKK6 inhibition leads to the reduced levels of the transcription factor SOX9. In line with this finding, deletion of SOX9 with CRISPR/Cas9 resulted in decreased proliferation of EACs in 3D organoid culture and reduced tumor growth. Together these findings establish a druggable axis that can be harnessed for therapeutic gain against EAC.
Collapse
Affiliation(s)
- Sijie Lin
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA; Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian 350025, PR China
| | - Kuancan Liu
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA; Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian 350025, PR China; Dong fang Hospital, Xiamen University, Fuzhou, Fujian 350025, PR China.
| | - Yongchun Zhang
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA
| | - Ming Jiang
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA
| | - Rong Lu
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA
| | - Christopher J Folts
- Department of Biomedical Genetics, University of Rochester, Rochester NY14642, USA
| | - Xia Gao
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA
| | - Mark D Noble
- Department of Biomedical Genetics, University of Rochester, Rochester NY14642, USA
| | - Tingting Zhao
- Dong fang Hospital, Xiamen University, Fuzhou, Fujian 350025, PR China
| | - Zhongren Zhou
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, MO63110, USA
| | - Xiaopeng Lan
- Institute for Laboratory Medicine, Fuzhou General Hospital, PLA, Fuzhou, Fujian 350025, PR China; Dong fang Hospital, Xiamen University, Fuzhou, Fujian 350025, PR China.
| | - Jianwen Que
- Division of Digestive and Liver Diseases and Center for Human Development, Department of Medicine, Columbia University, NY 10032, USA.
| |
Collapse
|
37
|
Chen W, Zhao W, Chen S, Zhang L, Guo Z, Wang L, Wang J, Wan Z, Hong Y, Yu L. Expression and correlation of MALAT1 and SOX9 in non-small cell lung cancer. CLINICAL RESPIRATORY JOURNAL 2018; 12:2284-2291. [PMID: 29896925 DOI: 10.1111/crj.12906] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/12/2018] [Accepted: 04/07/2018] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) is the most common cause of cancer-related deaths in the world. MALAT1 and SOX9 have important roles in tumour formation and development in several types of cancers. However, little is known about the function and co-relationship of these 2 factors in NSCLC in vivo. OBJECTIVES To explore the role of MALAT1 and SOX9 expression relationship, their clinical pathological characteristics and OS on NSCLC patients. METHODS Paired of primary lung cancer tissues and the matched tumour adjacent tissues were collected in 121 NSCLC patients. MALAT1 and SOX9 mRNA expression was measured by SYBR green q RT-PCR assay. SOX-9 protein expression was measured by streptavidin-peroxidase (SP) staining method. RESULTS MALAT1and SOX9 expression was higher in NSCLC tissues than the adjacent tissues, and they have positive correlation. Moreover, SOX9 protein expression was higher in NSCLC tissues, especially in MALAT1 mRNA higher expressed NSCLC tissues. MALAT1 and SOX9 mRNA expression were associated with age (x2 =11.474, P = .009), tumour size (x2 =26.839, P = .000), TNM stage (x2 =8.010, P = .046) and LEL. (x2 =53.908, P = .000). NSCLC patients with higher MALAT1 and SOX9 mRNA expression had poorer OS rates. CONCLUSIONS MALAT1 and SOX9 could be used as prognostic co-biomarker in NSCLC.
Collapse
Affiliation(s)
- Wei Chen
- Department of Respiratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Wei Zhao
- Department of Pathology, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sheng Chen
- Department of Pulmonary Surgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Li Zhang
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Zhongying Guo
- Department of Pathology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Lixin Wang
- Department of Respiratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Jipeng Wang
- Department of Respiratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Zongren Wan
- Department of Respiratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Yongqing Hong
- Department of Respiratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Liang Yu
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China.,Key Laboratory of Hematology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
38
|
Liang Y, Feng Y, Zong M, Wei X, Lee J, Feng Y, Li H, Yang G, Wu ZJ, Fu XD, Feng GS. β-catenin deficiency in hepatocytes aggravates hepatocarcinogenesis driven by oncogenic β-catenin and MET. Hepatology 2018; 67:1807-1822. [PMID: 29152756 PMCID: PMC5906147 DOI: 10.1002/hep.29661] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/30/2017] [Accepted: 11/13/2017] [Indexed: 12/28/2022]
Abstract
UNLABELLED Both activating and inactivating mutations in catenin β1 (ctnnb1), which encodes β-catenin, have been implicated in liver tumorigenesis in humans and mice, although the underlying mechanisms are not fully understood. Herein, we show that deletion of endogenous β-catenin in hepatocytes aggravated hepatocellular carcinoma (HCC) development driven by an oncogenic version of β-catenin (CAT) in combination with the hepatocyte growth factor receptor MET proto-oncogene receptor tyrosine kinase (MET). Although the mitogenic signaling and cell cycle progression was modestly impaired after CAT/MET transfection, the β-catenin-deficient livers displayed changes in transcriptomes, increased DNA damage response, expanded Sox9+ cells, and up-regulation of protumorigenic cytokines, including interleukin-6 and transforming growth factor β1. These events eventually exacerbated CAT/MET-driven hepatocarcinogenesis in β-catenin-deficient livers, featured by up-regulation of extracellular signal-regulated kinase (Erk), protein kinase B (Akt), and Wnt/β-catenin signaling and cyclin D1 expression. The resultant mouse tumors showed similar transcriptomes to human HCC samples with concomitant CTNNB1 mutations and MET overexpression. CONCLUSION These data argue that while dominantly activating mutants of β-catenin are oncogenic, inhibiting the oncogenic signaling pathway generates a pro-oncogenic microenvironment that may facilitate HCC recurrence following a targeted therapy of the primary tumor. An effective therapeutic strategy must require disruption of the oncogenic signaling in tumor cells and suppression of the secondary tumor-promoting stromal effects in the liver microenvironment. (Hepatology 2018;67:1807-1822).
Collapse
Affiliation(s)
- Yan Liang
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA
| | - Yun Feng
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA,The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Min Zong
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA
| | - Xufu Wei
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA,Department of Hepatology, 1 affiliated Hospital, Chong-Qing Medical University, China
| | - Jin Lee
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA
| | - Yukuan Feng
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA,Department of Anatomy, Mudanjiang Medical College, Mudanjiang, Heilongjiang 157011, China
| | - Hairi Li
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Guangshun Yang
- The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Zhong-Jun Wu
- Department of Hepatology, 1 affiliated Hospital, Chong-Qing Medical University, China
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Gen-Sheng Feng
- Department of Pathology, and Division of Biological Sciences, Moores UCSD Cancer Center, University of California San Diego, La Jolla, California 92093, USA, Corresponding to: Gen-Sheng Feng,
| |
Collapse
|
39
|
Zhang N, Chai D, Du H, Li K, Xie W, Li X, Yang R, Lian X, Xu Y. Expression of Reg IV and SOX9 and their correlation in human gastric cancer. BMC Cancer 2018; 18:344. [PMID: 29587675 PMCID: PMC5870489 DOI: 10.1186/s12885-018-4285-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/21/2018] [Indexed: 01/06/2023] Open
Abstract
Background Reg IV is a member of the regenerating gene family and has been demonstrated to be overexpressed in gastric cancer. However, the functional mechanism of Reg IV in gastric cancer is still unclear. Methods Expression of Reg IV and SOX9 were investigated by immunohistochemistry (IHC) and real-time PCR, and the correlation between the expression of Reg IV and SOX9 was analyzed in gastric cancer tissues. Reg IV expression vectors and a siRNA of Reg IV and SOX9 were transfected into human gastric cancer cells and the protein and mRNA levels of Reg IV and SOX9 were investigated by western blot and real-time PCR. The invasion and migration ability of gastric cancer cells with overexpressed Reg IV and with gene silence of Reg IV and SOX9 were examined by transwell chambers and wound healing assay. Results The Reg IV and SOX9 protein expression levels were both significantly higher in gastric cancer tissues compared with adjacent tissues (p = 0.022, p = 0.003). Reg IV protein expression significantly correlated with tumor invasion depth (p < 0.001), but had no significant correlations with age, clinical stage or lymph node metastasis. SOX9 protein expression also had no significant correlations with age, clinical stage, tumor invasion depth or lymph node metastasis. Reg IV transcript expression demonstrated a significant correlation with invasion depth and lymph node metastasis (p = 0.005, p < 0.001) and no significant correlations with age, clinical stage, tumor tissue differentiation or tumor size. SOX9 transcript expression demonstrated a significant correlation with invasion depth and tumor tissue differentiation (p = 0.044, p = 0.007) and no significant correlations with age, clinical stage or tumor size. The Reg IV expression showed a positive correlation with the SOX9 expression (p < 0.000, p = 0.008). Overexpression of Reg IV could upregulate SOX9 expression and promote invasiveness and migration of tumor cells, and silencing of Reg IV could downregulate SOX9 and inhibit invasiveness and migration of tumor cells in MKN-45 and AGS cells. On the other hand, silencing of SOX9 could upregulate Reg IV protein expression. Conclusions Our study demonstrated that Reg IV positively regulates the expression of SOX9 and is involved in tumor cell invasion and migration in gastric cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4285-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Na Zhang
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China.,College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Dandan Chai
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Huifen Du
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Kesheng Li
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China. .,College of Life Sciences, Lanzhou University, Lanzhou, China.
| | - Wenguang Xie
- College of Life Sciences, Lanzhou University, Lanzhou, China
| | - Xingwen Li
- Department of Surgery, Tumor Hospital of Gansu Province, Lanzhou, China
| | - Rong Yang
- Department of pathology, Tumor Hospital of Gansu Province, Lanzhou, China
| | - Xiaowen Lian
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| | - Yang Xu
- Department of Medicine Biotechnology, Medicine and Science Research Institute of Gansu Province, Lanzhou, China
| |
Collapse
|
40
|
Hatina J, Parmar HS, Kripnerova M, Hepburn A, Heer R. Urothelial Carcinoma Stem Cells: Current Concepts, Controversies, and Methods. Methods Mol Biol 2018; 1655:121-136. [PMID: 28889382 DOI: 10.1007/978-1-4939-7234-0_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cancer stem cells are defined as a self-renewing and self-protecting subpopulation of cancer cells able to differentiate into morphologically and functionally diverse cancer cells with a limited lifespan. To purify cancer stem cells, two basic approaches can be applied, the marker-based approach employing various more of less-specific cell surface marker molecules and a marker-free approach largely based on various self-protection mechanisms. Within the context of urothelial carcinoma, both methods could find use. The cell surface markers have been mainly derived from the urothelial basal cell, a probable cell of origin of muscle-invasive urothelial carcinoma, with CD14, CD44, CD90, and 67LR representing successful examples of this strategy. The marker-free approaches involve side population sorting, for which a detailed protocol is provided, as well as the Aldefluor assay, which rely on a specific overexpression of efflux pumps or the detoxification enzyme aldehyde dehydrogenase, respectively, in stem cells. These assays have been applied to both non-muscle-invasive and muscle-invasive bladder cancer samples and cell lines. Urothelial carcinoma stem cells feature a pronounced heterogeneity as to their molecular stemness mechanisms. Several aspects of urothelial cancer stem cell biology could enter translational development rather soon, e.g., a specific CD44+-derived gene expression signature able to identify non-muscle-invasive bladder cancer patients with a high risk of progression, or deciphering a mechanism responsible for repopulating activity of urothelial carcinoma stem cells within the context of therapeutic resistance.
Collapse
Affiliation(s)
- Jiri Hatina
- Faculty of Medicine in Pilsen, Institute of Biology, Charles University in Prague, Plzen, Czech Republic.
| | - Hamendra Singh Parmar
- Faculty of Medicine in Pilsen, Institute of Biology, Charles University in Prague, Plzen, Czech Republic
| | - Michaela Kripnerova
- Faculty of Medicine in Pilsen, Institute of Biology, Charles University in Prague, Plzen, Czech Republic
| | - Anastasia Hepburn
- Solid Tumour Target Discovery Laboratory, Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | - Rakesh Heer
- Solid Tumour Target Discovery Laboratory, Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| |
Collapse
|
41
|
Wang Z, Xu X, Liu N, Cheng Y, Jin W, Zhang P, Wang X, Yang H, Liu H, Tu Y. SOX9-PDK1 axis is essential for glioma stem cell self-renewal and temozolomide resistance. Oncotarget 2017; 9:192-204. [PMID: 29416606 PMCID: PMC5787456 DOI: 10.18632/oncotarget.22773] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive brain tumor with limited therapeutic options. Glioma stem cell (GSC) is thought to be greatly responsible for glioma tumor progression and drug resistance. But the molecular mechanisms of GSC deriving recurrence and drug resistance are still unclear. SOX9 (sex-determining region Y (SRY)-box9 protein), a transcription factor expressed in most solid tumors, is reported as a key regulator involved in maintaining cancer hallmarks including the GSCs state. Previously, we have observed that silencing of SOX9 suppressed glioma cells proliferation both in vitro and in vivo. Here, we found that SOX9 was essential for GSC self-renewal. Silencing of SOX9 down-regulated a broad range of stem cell markers and inhibited glioma cell colony and sphere formation. We identified pyruvate dehydrogenase kinase 1 (PDK1) as a target gene of SOX9 using microarray analyses. PDK1 inactivation greatly inhibited glioma cell colony and sphere formation and sensitized glioma spheres to temozolomide (TMZ) toxicity. In addition, SOX9-shRNA and PDK1 inhibitor could greatly sensitize GSC to TMZ in vivo. Taken together, our data reveals that SOX9-PDK1 axis is a key regulator of GSC self-renewal and GSC temozolomide resistance. These findings may provide help for future human GBM therapy.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China.,Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiao Tong University, Xi'an 710049, China
| | - Xiaoshan Xu
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China
| | - Nan Liu
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China
| | - Yingduan Cheng
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China.,Department of Research, Cipher Ground, North Brunswick, NJ 08902, USA
| | - Weilin Jin
- Institute of Nano Biomedicine and Engineering, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Pengxing Zhang
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hongwei Yang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hui Liu
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China
| | - Yanyang Tu
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical, University, Xi'an 710038, China.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
42
|
Loss of Extracellular Signal-Regulated Kinase 1/2 in the Retinal Pigment Epithelium Leads to RPE65 Decrease and Retinal Degeneration. Mol Cell Biol 2017; 37:MCB.00295-17. [PMID: 29038159 PMCID: PMC5705814 DOI: 10.1128/mcb.00295-17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/02/2017] [Indexed: 12/19/2022] Open
Abstract
Recent work suggested that the activity of extracellular signal-regulated kinase 1/2 (ERK1/2) is increased in the retinal pigment epithelium (RPE) of age-related macular degeneration (ARMD) patients and therefore could be an attractive therapeutic target. Notably, ERK1/2 pathway inhibitors are used in cancer therapy, with severe and noncharacterized ocular side effects. To decipher the role of ERK1/2 in RPE cells, we conditionally disrupted the Erk1 and Erk2 genes in mouse RPE. The loss of ERK1/2 activity resulted in a significant decrease in the level of RPE65 expression, a decrease in ocular retinoid levels concomitant with low visual function, and a rapid disorganization of RPE cells, ultimately leading to retinal degeneration. Our results identify the ERK1/2 pathway as a direct regulator of the visual cycle and a critical component of the viability of RPE and photoreceptor cells. Moreover, our results caution about the need for a very fine adjustment of kinase inhibition in cancer or ARMD treatment in order to avoid ocular side effects.
Collapse
|
43
|
Li H, Cai H, Deng J, Tu X, Sun Y, Huang Z, Ding Z, Dong L, Chen J, Zang Y, Zhang J. TGF-β-mediated upregulation of Sox9 in fibroblast promotes renal fibrosis. Biochim Biophys Acta Mol Basis Dis 2017; 1864:520-532. [PMID: 29158184 DOI: 10.1016/j.bbadis.2017.11.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 10/26/2017] [Accepted: 11/16/2017] [Indexed: 10/18/2022]
Abstract
TGF-β signaling plays a principal role in renal fibrosis, but the precise mechanisms and the downstream factors are still largely unknown. Sox9 exhibits diverse roles in regulating the production of extracellular matrix proteins. Here we found that Sox9 was induced by TGF-β in the kidney fibroblast and acted as an important downstream mediator of TGF-β signaling in promoting renal fibrosis. TGF-β/Smad signaling mediated the upregulation of Sox9 in kidney fibroblast by binding to a conserved enhancer. In different mouse models of renal fibrosis, as well as in the kidney biopsy tissue from patients with renal fibrosis, Sox9 expression significantly increased. Immunostaining confirmed the upregulation of Sox9 in the kidney fibroblast during renal fibrosis. Delivery of Sox9 knockdown plasmid to the kidney by ultrasound microbubble-mediated gene transfer suppressed the unilateral ureteral obstruction (UUO) or folic acid-induced mouse renal fibrosis, whereas ectopic expression of Sox9 aggravated renal fibrosis. In addition, we identified Sox9 as a direct target of miR-30. Notably, miR-30 expression was significantly inhibited by TGF-β1 in the kidney fibroblast and the downregulation of miR-30 was observed in renal fibrosis. Mechanistically, inhibition of miR-30 independently strengthened the effect of TGF-β/Smad signaling on Sox9 upregulation. Adenovirus-mediated ectopic expression of miR-30 in kidney fibroblast greatly reduced UUO-induced renal fibrosis by targeting Sox9. These findings link Sox9 to intrinsic mechanisms of TGF-β signaling in renal fibrosis and may have therapeutic potential for tissue fibrosis.
Collapse
Affiliation(s)
- Huanan Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Huimin Cai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Jia Deng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Xiaolong Tu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Yanyan Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Zhi Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China.
| | - Yuhui Zang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China.
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, PR China; Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing 210093, PR China.
| |
Collapse
|
44
|
Azar S, Leventoux N, Ripoll C, Rigau V, Gozé C, Lorcy F, Bauchet L, Duffau H, Guichet PO, Rothhut B, Hugnot JP. Cellular and molecular characterization of IDH1-mutated diffuse low grade gliomas reveals tumor heterogeneity and absence of EGFR/PDGFRα activation. Glia 2017; 66:239-255. [PMID: 29027701 DOI: 10.1002/glia.23240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/14/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022]
Abstract
Diffuse low grade gliomas (DLGG, grade II gliomas) are slowly-growing brain tumors that often progress into high grade gliomas. Most tumors have a missense mutation for IDH1 combined with 1p19q codeletion in oligodendrogliomas or ATRX/TP53 mutations in astrocytomas. The phenotype of tumoral cells, their environment and the pathways activated in these tumors are still ill-defined and are mainly based on genomics and transcriptomics analysis. Here we used freshly-resected tumors to accurately characterize the tumoral cell population and their environment. In oligodendrogliomas, cells express the transcription factors MYT1, Nkx2.2, Olig1, Olig2, Sox8, four receptors (EGFR, PDGFRα, LIFR, PTPRZ1) but not the co-receptor NG2 known to be expressed by oligodendrocyte progenitor cells. A variable fraction of cells also express the more mature oligodendrocytic markers NOGO-A and MAG. DLGG cells are also stained for the young-neuron marker doublecortin (Dcx) which is also observed in oligodendrocytic cells in nontumoral human brain. In astrocytomas, MYT1, PDGFRα, PTPRZ1 were less expressed whereas Sox9 was prominent over Sox8. The phenotype of DLGG cells is overall maintained in culture. Phospho-array screening showed the absence of EGFR and PDGFRα phosphorylation in DLGG but revealed the strong activation of p44/42 MAPK/ERK which was present in a fraction of tumoral cells but also in nontumoral cells. These results provide evidence for the existence of close relationships between the cellular phenotype and the mutations found in DLGG. The slow proliferation of these tumors may be associated with the absence of activation of PDGFRα/EGFR receptors.
Collapse
Affiliation(s)
- S Azar
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - N Leventoux
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - C Ripoll
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - V Rigau
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - C Gozé
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Genetics Department, Hôpital Gui de Chauliac, Montpellier, France
| | - F Lorcy
- CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - L Bauchet
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Surgery Department, Hôpital Gui de Chauliac, Montpellier, France
| | - H Duffau
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Surgery Department, Hôpital Gui de Chauliac, Montpellier, France
| | - P O Guichet
- LNEC Inserm U1084 1 rue Georges Bonnet 86022 Poitiers Cedex, France
| | - B Rothhut
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - J P Hugnot
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,University of Montpellier, Faculty of Sciences, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| |
Collapse
|
45
|
Small bowel carcinomas in celiac or Crohn's disease: distinctive histophenotypic, molecular and histogenetic patterns. Mod Pathol 2017; 30:1453-1466. [PMID: 28664941 DOI: 10.1038/modpathol.2017.40] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/25/2022]
Abstract
Non-familial small bowel carcinomas are relatively rare and have a poor prognosis. Two small bowel carcinoma subsets may arise in distinct immune-inflammatory diseases (celiac disease and Crohn's disease) and have been recently suggested to differ in prognosis, celiac disease-associated carcinoma cases showing a better outcome, possibly due to their higher DNA microsatellite instability and tumor-infiltrating T lymphocytes. In this study, we investigated the histological structure (glandular vs diffuse/poorly cohesive, mixed or solid), cell phenotype (intestinal vs gastric/pancreatobiliary duct type) and Wnt signaling activation (β-catenin and/or SOX-9 nuclear expression) in a series of 26 celiac disease-associated small bowel carcinoma, 25 Crohn's disease-associated small bowel carcinoma and 25 sporadic small bowel carcinoma cases, searching for new prognostic parameters. In addition, non-tumor mucosa of celiac and Crohn's disease patients was investigated for epithelial precursor changes (hyperplastic, metaplastic or dysplastic) to help clarify carcinoma histogenesis. When compared with non-glandular structure and non-intestinal phenotype, both glandular structure and intestinal phenotype were associated with a more favorable outcome at univariable or stage- and microsatellite instability/tumor-infiltrating lymphocyte-inclusive multivariable analysis. The prognostic power of histological structure was independent of the clinical groups while the non-intestinal phenotype, associated with poor outcome, was dominant among Crohn's disease-associated carcinoma. Both nuclear β-catenin and SOX-9 were preferably expressed among celiac disease-associated carcinomas; however, they were devoid, per se, of prognostic value. We obtained findings supporting an origin of celiac disease-associated carcinoma in SOX-9-positive immature hyperplastic crypts, partly through flat β-catenin-positive dysplasia, and of Crohn's disease-associated carcinoma in a metaplastic (gastric and/or pancreatobiliary-type) mucosa, often through dysplastic polypoid growths of metaplastic phenotype. In conclusion, despite their common origin in a chronically inflamed mucosa, celiac disease-associated and Crohn's disease-associated small bowel carcinomas differ substantially in histological structure, phenotype, microsatellite instability/tumor-infiltrating lymphocyte status, Wnt pathway activation, mucosal precursor lesions and prognosis.
Collapse
|
46
|
Liu J, Kumar S, Dolzhenko E, Alvarado GF, Guo J, Lu C, Chen Y, Li M, Dessing MC, Parvez RK, Cippà PE, Krautzberger AM, Saribekyan G, Smith AD, McMahon AP. Molecular characterization of the transition from acute to chronic kidney injury following ischemia/reperfusion. JCI Insight 2017; 2:94716. [PMID: 28931758 PMCID: PMC5612583 DOI: 10.1172/jci.insight.94716] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/10/2017] [Indexed: 12/16/2022] Open
Abstract
Though an acute kidney injury (AKI) episode is associated with an increased risk of chronic kidney disease (CKD), the mechanisms determining the transition from acute to irreversible chronic injury are not well understood. To extend our understanding of renal repair, and its limits, we performed a detailed molecular characterization of a murine ischemia/reperfusion injury (IRI) model for 12 months after injury. Together, the data comprising RNA-sequencing (RNA-seq) analysis at multiple time points, histological studies, and molecular and cellular characterization of targeted gene activity provide a comprehensive profile of injury, repair, and long-term maladaptive responses following IRI. Tubular atrophy, interstitial fibrosis, inflammation, and development of multiple renal cysts were major long-term outcomes of IRI. Progressive proximal tubular injury tracks with de novo activation of multiple Krt genes, including Krt20, a biomarker of renal tubule injury. RNA-seq analysis highlights a cascade of temporal-specific gene expression patterns related to tubular injury/repair, fibrosis, and innate and adaptive immunity. Intersection of these data with human kidney transplant expression profiles identified overlapping gene expression signatures correlating with different stages of the murine IRI response. The comprehensive characterization of incomplete recovery after ischemic AKI provides a valuable resource for determining the underlying pathophysiology of human CKD.
Collapse
Affiliation(s)
- Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Egor Dolzhenko
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Can Lu
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Meng Li
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Mark C Dessing
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - A Michaela Krautzberger
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, California, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
47
|
Bohuslavova R, Cerychova R, Nepomucka K, Pavlinkova G. Renal injury is accelerated by global hypoxia-inducible factor 1 alpha deficiency in a mouse model of STZ-induced diabetes. BMC Endocr Disord 2017; 17:48. [PMID: 28774305 PMCID: PMC5543752 DOI: 10.1186/s12902-017-0200-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hypoxia inducible factor 1 (HIF-1) activates protective pathways to counteract hypoxia and prevent tissue damage in conjunction with renal injury. The aim of this study was to evaluate a role of HIF-1 in diabetes-induced kidney damage. METHODS We used a streptozotocin-induced diabetes mouse model and compared biochemical, histological and molecular parameters associated with kidney damage in Hif1α deficient (Hif1α +/- ) and wild-type mice. RESULTS We showed that Hif1α deficiency accelerated pathological changes in the early stage of DN. Six weeks after diabetes-induction, Hif1α deficient mice showed more prominent changes in biochemical serum parameters associated with glomerular injury, increased expression of podocyte damage markers, and loss of podocytes compared to wild-type mice. These results indicate that Hif1α deficiency specifically affects podocyte survival in the early phase of DN, resulting in diabetic glomerular injury. In contrast, renal fibrosis was not affected by the global reduction of Hif1α, at least not in the early phase of diabetic exposure. CONCLUSIONS Together our data reveal that HIF-1 has an essential role in the early response to prevent diabetes-induced tissue damage and that impaired HIF-1 signaling results in a faster progression of DN. Although the modulation of HIF-1 activity is a high-priority target for clinical treatments, further study is required to investigate HIF-1 as a potential therapeutic target for the treatment of DN.
Collapse
Affiliation(s)
- Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, Vestec, 25242 Czechia
| | - Radka Cerychova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, Vestec, 25242 Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Katerina Nepomucka
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, Vestec, 25242 Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology CAS, BIOCEV, Center of Excellence, Prumyslova 595, Vestec, 25242 Czechia
| |
Collapse
|
48
|
Chen H, Garbutt CC, Spentzos D, Choy E, Hornicek FJ, Duan Z. Expression and Therapeutic Potential of SOX9 in Chordoma. Clin Cancer Res 2017; 23:5176-5186. [PMID: 28606919 DOI: 10.1158/1078-0432.ccr-17-0177] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/21/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Conventional chemotherapeutic agents are ineffective in the treatment of chordoma. We investigated the functional roles and therapeutic relevance of the sex-determining region Y (SRY)-box 9 (SOX9) in chordoma.Experimental Design: SOX9 expression was examined by immunohistochemistry (IHC) using 50 chordoma tissue samples. SOX9 expression in chordoma cell lines was examined by Western blot and immunofluorescent assays. We used synthetic human SOX9 siRNA to inhibit the expression of SOX9. Cell proliferation ability and cytotoxicity of inhibiting SOX9 were assessed by 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide (MTT) and clonogenic assays. The effect of SOX9 knockdown on chordoma cell motility was evaluated by a wound-healing assay and a Transwell invasion chamber assay. Knockdown of SOX9 induced apoptosis, cell-cycle arrest, as well as decreased expression of cancer stem cell markers were determined by Western blot and flow cytometric assays. The effect of the combination of SOX9 siRNA and the chemotherapeutic drug doxorubicin/cisplatin on chordoma cells was assessed by an MTT assay.Results: Tissue microarray and IHC analysis showed that SOX9 is broadly expressed in chordomas and that higher expression levels of SOX9 correlated with a poor prognosis. RNA interference (RNAi)-mediated knockdown of SOX9 inhibited chordoma cell growth, decreased cell motility, and induced apoptosis as well as cell-cycle arrest. Moreover, the combination of SOX9 inhibition and chemotherapeutic drugs had an enhanced anti-cancer effect on chordoma cells.Conclusions: Our results demonstrate that SOX9 plays a crucial role in chordoma. Targeting SOX9 provides a new rationale for treatment of chordoma. Clin Cancer Res; 23(17); 5176-86. ©2017 AACR.
Collapse
Affiliation(s)
- Hua Chen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Emergency Surgery, ShenZhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, Guangdong Province, China
| | - Cassandra C Garbutt
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dimitrios Spentzos
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
49
|
Balsara ZR, Li X. Sleeping beauty: awakening urothelium from its slumber. Am J Physiol Renal Physiol 2017; 312:F732-F743. [PMID: 28122714 PMCID: PMC5407074 DOI: 10.1152/ajprenal.00337.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 12/18/2022] Open
Abstract
The bladder urothelium is essentially quiescent but regenerates readily upon injury. The process of urothelial regeneration harkens back to the process of urothelial development whereby urothelial stem/progenitor cells must proliferate and terminally differentiate to establish all three urothelial layers. How the urothelium regulates the level of proliferation and the timing of differentiation to ensure the precise degree of regeneration is of significant interest in the field. Without a carefully-orchestrated process, urothelial regeneration may be inadequate, thereby exposing the host to toxins or pathogens. Alternatively, regeneration may be excessive, thereby setting the stage for tumor development. This review describes our current understanding of urothelial regeneration. The current controversies surrounding the identity and location of urothelial progenitor cells that mediate urothelial regeneration are discussed and evidence for each model is provided. We emphasize the factors that have been shown to be crucial for urothelial regeneration, including local growth factors that stimulate repair, and epithelial-mesenchymal cross talk, which ensures feedback regulation. Also highlighted is the emerging concept of epigenetic regulation of urothelial regeneration, which additionally fine tunes the process through transcriptional regulation of cell cycle genes and growth and differentiation factors. Finally, we emphasize how several of these pathways and/or programs are often dysregulated during malignant transformation, further corroborating their importance in directing normal urothelial regeneration. Together, evidence in the field suggests that any attempt to exploit regenerative programs for the purposes of enhanced urothelial repair or replacement must take into account this delicate balance.
Collapse
Affiliation(s)
- Zarine R Balsara
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; and
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xue Li
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; and
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Popp FC, Popp MC, Zhao Y, Betzler C, Kropf S, Garlipp B, Benckert C, Kalinski T, Lippert H, Bruns CJ. Protocol of the PANCALYZE trial: a multicenter, prospective study investigating the tumor biomarkers CXCR4, SMAD4, SOX9 and IFIT3 in patients with resected pancreatic adenocarcinoma to predict the pattern of recurrence of the disease. BMC Cancer 2017; 17:229. [PMID: 28356064 PMCID: PMC5371262 DOI: 10.1186/s12885-017-3186-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 03/09/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies today with an urgent need for novel therapeutic strategies. Biomarker analysis helps to better understand tumor biology and might emerge as a tool to develop personalized therapies. The aim of the study is to investigate four promising biomarkers to predict the clinical course and particularly the pattern of tumor recurrence after surgical resection. DESIGN Patients undergoing surgery for PDAC can be enrolled into the PANCALYZE trial. Biomarker expression of CXCR4, SMAD4, SOX9 and IFIT3 will be prospectively assessed by immunohistochemistry and verified by rt.-PCR from tumor and adjacent healthy pancreatic tissue of surgical specimen. Immunohistochemistry expression pattern of all four biomarkers will be combined into a single score. Beginning with the hospital stay clinical data from enrolled patients will be collected and followed. Different adjuvant chemotherapy protocols will be used to create subgroups. The combined biomarker expression score will be correlated with the further clinical course of the patients to test the hypothesis if CXCR4 positive, SMAD4 negative, SOX9 positive, IFIT3 positive tumors will predominantly develop metastatic spread. DISCUSSION Pancreatic cancer is associated with different patterns of progression requiring personalized therapeutic strategies. Biomarker expression analysis might be a tool to predict the pattern of tumor recurrence and discriminate patients that develop systemic metastatic disease from those with tumors that rather develop local recurrence over time. This data might lead to personalized adjuvant treatment decisions as patients with tumors that stay localized might benefit from adjuvant local therapies like radiochemotherapy as compared to those with systemic recurrence who would benefit exclusively from chemotherapy. Moreover, the pattern of propagation might be a predefined characteristic of pancreatic cancer determined by the genetic signature of the tumor. In the future, biomarker expression analysis could be performed on tumor biopsies to develop personalized therapeutic pathways right after diagnosis of cancer. TRIAL REGISTRATION German Clinical Trials Register, DRKS00006179 .
Collapse
Affiliation(s)
- Felix C. Popp
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Marie Christine Popp
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Yue Zhao
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christopher Betzler
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Siegfried Kropf
- Institute for Biometrics and Medical Informatics, Otto-von-Guericke University, Magdeburg, Germany
| | - Benjamin Garlipp
- Clinic for General, Visceral and Vascular Surgery, University Clinic Magdeburg, Magdeburg, Germany
| | - Christoph Benckert
- Department of General and Visceral Surgery, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Thomas Kalinski
- Department of Pathology, Otto von Guericke University, Magdeburg, Germany
| | - Hans Lippert
- Institute for Quality Control in Operative Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Christiane J. Bruns
- Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| |
Collapse
|