1
|
Manrique-Caballero CL, Barasch J, Zaidi SK, Bates CM, Ray EC, Kleyman TR, Al-Bataineh MM. Expression and distribution of MUC1 in the developing and adult kidney. Am J Physiol Renal Physiol 2025; 328:F107-F120. [PMID: 39588770 PMCID: PMC11918333 DOI: 10.1152/ajprenal.00206.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
Mucin 1 (or MUC1) is a heterodimeric transmembrane glycoprotein expressed on the apical surface of polarized epithelial cells in several tissues including the kidney. Recent studies have revealed several novel roles of MUC1 in the kidney, potentially including bacterial infection, mineral balance, and genetic interstitial kidney disease, even though MUC1 levels are reduced not only in the kidney but also in all tissues due to MUC1 mutations. A careful localization of MUC1 in discrete segments of the nephron is the first step in understanding the multiple functional roles of MUC1 in the kidney. Most published reports of MUC1 expression to date have been largely confined to cultured cells, tumor tissues, and selective nephron segments of experimental rodents, and very few studies have been performed using human kidney tissues. Given the rising attention to the role of MUC1 in differing components of renal physiology, we carefully examined the kidney distribution of MUC1 in both human and mouse kidney sections using well-defined markers for different nephron segments or cell types. We further extended our investigation to include sections of early stages of mouse kidney development and upon injury in humans. We included staining for MUC1 in urothelial cells, the highly specialized epithelial cells lining the renal pelvis and bladder. These data implicate a role for MUC1 in antimicrobial defense. Our study provides the groundwork to test the physiological relevance of MUC1 in the urinary tract.NEW & NOTEWORTHY MUC1 is a transmembrane glycoprotein expressed on the apical surface of polarized epithelial tissues and most carcinomas. MUC1 may play novel roles in the kidney including defense against infections. Here, we examine the expression of MUC1 in mouse and human kidneys. We show that the distal nephron and the urinary system are the predominant sites of expression of both message and protein, implicating segment-specific roles including distal nephron defense against ascending bacteria.
Collapse
Affiliation(s)
- Carlos L Manrique-Caballero
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States
| | - Syed K Zaidi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Carlton M Bates
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Evan C Ray
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Mohammad M Al-Bataineh
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
2
|
Mao W, Zhang H, Wang K, Geng J, Wu J. Research progress of MUC1 in genitourinary cancers. Cell Mol Biol Lett 2024; 29:135. [PMID: 39491020 PMCID: PMC11533421 DOI: 10.1186/s11658-024-00654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
MUC1 is a highly glycosylated transmembrane protein with a high molecular weight. It plays a role in lubricating and protecting mucosal epithelium, participates in epithelial cell renewal and differentiation, and regulates cell adhesion, signal transduction, and immune response. MUC1 is expressed in both normal and malignant epithelial cells, and plays an important role in the diagnosis, prognosis prediction and clinical monitoring of a variety of tumors and is expected to be a new therapeutic target. This article reviews the structural features, expression regulation mechanism, and research progress of MUC1 in the development of genitourinary cancers and its clinical applications.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| | - Houliang Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Keyi Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Jiang Geng
- Department of Urology, Bengbu First People's Hospital, Bengbu, People's Republic of China.
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Jianping Wu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
3
|
Medina-Dols A, Cañellas G, Capó T, Solé M, Mola-Caminal M, Cullell N, Jaume M, Nadal-Salas L, Llinàs J, Gómez L, Tur S, Jiménez C, Díaz RM, Carrera C, Muiño E, Gallego-Fabrega C, Soriano-Tárraga C, Ruiz-Guerra L, Pol-Fuster J, Asensio V, Muncunill J, Fleischer A, Iglesias A, Giralt-Steinhauer E, Lazcano U, Fernández-Pérez I, Jiménez-Balado J, Gabriel-Salazar M, Garcia-Gabilondo M, Lei T, Torres-Aguila NP, Cárcel-Márquez J, Lladó J, Olmos G, Rosell A, Montaner J, Planas AM, Rabionet R, Hernández-Guillamon M, Jiménez-Conde J, Fernández-Cadenas I, Vives-Bauzá C. Role of PATJ in stroke prognosis by modulating endothelial to mesenchymal transition through the Hippo/Notch/PI3K axis. Cell Death Discov 2024; 10:85. [PMID: 38368420 PMCID: PMC10874379 DOI: 10.1038/s41420-024-01857-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024] Open
Abstract
Through GWAS studies we identified PATJ associated with functional outcome after ischemic stroke (IS). The aim of this study was to determine PATJ role in brain endothelial cells (ECs) in the context of stroke outcome. PATJ expression analyses in patient's blood revealed that: (i) the risk allele of rs76221407 induces higher expression of PATJ, (ii) PATJ is downregulated 24 h after IS, and (iii) its expression is significantly lower in those patients with functional independence, measured at 3 months with the modified Rankin scale ((mRS) ≤2), compared to those patients with marked disability (mRS = 4-5). In mice brains, PATJ was also downregulated in the injured hemisphere at 48 h after ischemia. Oxygen-glucose deprivation and hypoxia-dependent of Hypoxia Inducible Factor-1α also caused PATJ depletion in ECs. To study the effects of PATJ downregulation, we generated PATJ-knockdown human microvascular ECs. Their transcriptomic profile evidenced a complex cell reprogramming involving Notch, TGF-ß, PI3K/Akt, and Hippo signaling that translates in morphological and functional changes compatible with endothelial to mesenchymal transition (EndMT). PATJ depletion caused loss of cell-cell adhesion, upregulation of metalloproteases, actin cytoskeleton remodeling, cytoplasmic accumulation of the signal transducer C-terminal transmembrane Mucin 1 (MUC1-C) and downregulation of Notch and Hippo signaling. The EndMT phenotype of PATJ-depleted cells was associated with the nuclear recruitment of MUC1-C, YAP/TAZ, β-catenin, and ZEB1. Our results suggest that PATJ downregulation 24 h after IS promotes EndMT, an initial step prior to secondary activation of a pro-angiogenic program. This effect is associated with functional independence suggesting that activation of EndMT shortly after stroke onset is beneficial for stroke recovery.
Collapse
Affiliation(s)
- Aina Medina-Dols
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
| | - Guillem Cañellas
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Toni Capó
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Mola-Caminal
- Neurology, Hospital del Mar Medical Research Institute, Barcelona, Spain
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Natalia Cullell
- Neurology, Hospital Universitari Mútua de Terrassa/Fundacio Docència i Recerca Mútua Terrassa, Terrassa, Spain
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Marina Jaume
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Laura Nadal-Salas
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Jaume Llinàs
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Lluis Gómez
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Silvia Tur
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Neurology, Hospital Universitari Son Espases (HUSE), Palma, Spain
| | - Carmen Jiménez
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Neurology, Hospital Universitari Son Espases (HUSE), Palma, Spain
| | - Rosa M Díaz
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Neurology, Hospital Universitari Son Espases (HUSE), Palma, Spain
| | - Caty Carrera
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | | | - Laura Ruiz-Guerra
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
| | - Josep Pol-Fuster
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Víctor Asensio
- Department of Genetics (GEN-IB), HUSE, IdISBa, Palma, Spain
| | | | | | - Amanda Iglesias
- Department of Respiratory Medicine,, Hospital Universitari Son Espases-IdISBa Palma, Spain; CIBERES, Instituto de Salud Carlos III, Madrid, Spain
- CIBER of Respiratory Diseases (CIBERES), Madrid, Spain
| | | | - Uxue Lazcano
- Neurology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | | | | | - Marina Gabriel-Salazar
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Garcia-Gabilondo
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ting Lei
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Nuria-Paz Torres-Aguila
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Jerònia Lladó
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Gabriel Olmos
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Institute of Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville & Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Anna M Planas
- Department of Neuroscience and Experimental Therapeutics, Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Area of Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Raquel Rabionet
- Department of Genetics, Microbiology & Statistics, IBUB, University of Barcelona (UB), Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - Cristòfol Vives-Bauzá
- Neurobiology Laboratory, Research Unit, Hospital Universitari Son Espases, Health Research Institute of Balearic Islands (IdISBa), Palma, Spain.
- Department of Biology, University of Balearic Islands (UIB), Institut Universitari d'Investigacions en Ciències de la Salut (IUNICS), Palma, Spain.
| |
Collapse
|
4
|
Bourdon E, Swierczewski T, Goujon M, Boukrout N, Fellah S, Van der Hauwaert C, Larrue R, Lefebvre B, Van Seuningen I, Cauffiez C, Pottier N, Perrais M. MUC1 Drives the Progression and Chemoresistance of Clear Cell Renal Carcinomas. Cancers (Basel) 2024; 16:391. [PMID: 38254882 PMCID: PMC10814283 DOI: 10.3390/cancers16020391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
While the transmembrane glycoprotein mucin 1 (MUC1) is clustered at the apical borders of normal epithelial cells, with transformation and loss of polarity, MUC1 is found at high levels in the cytosol and is uniformly distributed over the entire surface of carcinoma cells, where it can promote tumor progression and adversely affects the response to therapy. Clear cell renal cell carcinoma (ccRCC), the main histotype of kidney cancer, is typically highly resistant to conventional and targeted therapies for reasons that remain largely unknown. In this context, we investigated whether MUC1 also plays a pivotal role in the cellular and molecular events driving ccRCC progression and chemoresistance. We showed, using loss- and gain-of-function approaches in ccRCC-derived cell lines, that MUC1 not only influences tumor progression but also induces a multi-drug-resistant profile reminiscent of the activation of ABC drug efflux transporters. Overall, our results suggest that targeting MUC1 may represent a novel therapeutic approach to limit ccRCC progression and improve drug sensitivity.
Collapse
Affiliation(s)
- Emma Bourdon
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Thomas Swierczewski
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Marine Goujon
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Nihad Boukrout
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Sandy Fellah
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Cynthia Van der Hauwaert
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Romain Larrue
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
- CHU Lille, Service de Toxicologie et Génopathies, F-59000 Lille, France
| | - Bruno Lefebvre
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR-S1172, Neuroscience & Cognition, Alzheimer & Tauopathies, F-59000 Lille, France;
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Christelle Cauffiez
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| | - Nicolas Pottier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR-S1172, Neuroscience & Cognition, Alzheimer & Tauopathies, F-59000 Lille, France;
| | - Michaël Perrais
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277–CANTHER–Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (E.B.); (T.S.); (M.G.); (N.B.); (S.F.); (C.V.d.H.); (R.L.); (I.V.S.); (C.C.); (N.P.)
| |
Collapse
|
5
|
Lu H, Xu WL, Wu YF, Yang WJ, Liu SW. Mucin 1 promotes salivary gland cancer cell proliferation and metastasis by regulating the epidermal growth factor receptor signaling pathway. J Dent Sci 2023; 18:1651-1662. [PMID: 37799896 PMCID: PMC10547961 DOI: 10.1016/j.jds.2023.01.035] [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: 01/20/2023] [Revised: 01/29/2023] [Indexed: 02/12/2023] Open
Abstract
Background/purpose Salivary gland cancer (SGC) is the common malignant tumor of the head and neck region with poor prognosis. Mucin 1 (MUC1) has been reported to be associated with the development of cancer. However, whether MUC1 contributed to the progression of SGC remains to be explored. Materials and methods Immunohistochemical analysis was used to explore the expression levels of MUC1 in SGC tissues. Cell proliferation, colony formation, wound healing, transwell, and xenograft assays were performed to examine the effects of MUC1 on SGC in vitro and in vivo. Results We found that the expression level of MUC1 was significantly upregulated in SGC tissues, and the expression level of MUC1 was significantly correlated with lymph node metastasis and TNM stage of SGC. Further exploration demonstrated that MUC1 knockdown drastically inhibited, while its overexpression promoted, cell growth, colony formation, migration, and invasion abilities of SGC cells in vitro. MUC1 knockdown significantly inhibited tumor growth in vivo, and vice versa. More importantly, we found that MUC1 promotes malignant phenotypes of SGC cells by regulating the epidermal growth factor receptor (EGFR) signaling pathway. Conclusion Our results revealed that MUC1 promotes the development of SGC by mediating the EGFR signaling pathway, which highlights the potential therapeutic target of MUC1/ EGFR in SGC.
Collapse
Affiliation(s)
- Hao Lu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wan-Lin Xu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Fan Wu
- Department of Oral Pathology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Jun Yang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Sheng-Wen Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
6
|
Jin W, Zhang M, Dong C, Huang L, Luo Q. The multifaceted role of MUC1 in tumor therapy resistance. Clin Exp Med 2023; 23:1441-1474. [PMID: 36564679 DOI: 10.1007/s10238-022-00978-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic resistances are frequently linked to the recurrence and poor prognosis of cancers and have been a key bottleneck in clinical tumor treatment. Mucin1 (MUC1), a heterodimeric transmembrane glycoprotein, exhibits abnormally overexpression in a variety of human tumors and has been confirmed to be related to the formation of therapeutic resistance. In this review, the multifaceted roles of MUC1 in tumor therapy resistance are summarized from aspects of pan-cancer principles shared among therapies and individual mechanisms dependent on different therapies. Concretely, the common mechanisms of therapy resistance across cancers include interfering with gene expression, promoting genome instability, modifying tumor microenvironment, enhancing cancer heterogeneity and stemness, and activating evasion and metastasis. Moreover, the individual mechanisms of therapy resistance in chemotherapy, radiotherapy, and biotherapy are introduced. Last but not least, MUC1-involved therapy resistance in different types of cancers and MUC1-related clinical trials are summarized.
Collapse
Affiliation(s)
- Weiqiu Jin
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mengwei Zhang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Changzi Dong
- Department of Bioengineering, School of Engineering and Science, University of Pennsylvania, Philadelphia, 19104, USA
| | - Lei Huang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
7
|
Zivotic M, Kovacevic S, Nikolic G, Mioljevic A, Filipovic I, Djordjevic M, Jovicic V, Topalovic N, Ilic K, Radojevic Skodric S, Dundjerovic D, Nesovic Ostojic J. SLUG and SNAIL as Potential Immunohistochemical Biomarkers for Renal Cancer Staging and Survival. Int J Mol Sci 2023; 24:12245. [PMID: 37569620 PMCID: PMC10418944 DOI: 10.3390/ijms241512245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Renal cell carcinoma (RCC) is the deadliest urological neoplasm. Up to date, no validated biomarkers are included in clinical guidelines for the screening and follow up of patients suffering from RCC. Slug (Snail2) and Snail (Snail1) belong to the Snail superfamily of zinc finger transcriptional factors that take part in the epithelial-mesenchymal transition, a process important during embryogenesis but also involved in tumor progression. We examined Slug and Snail immunohistochemical expression in patients with different stages of renal cell carcinomas with the aim to investigate their potential role as staging and prognostic factors. A total of 166 samples of malignant renal cell neoplasms were analyzed using tissue microarray and immunohistochemistry. Slug and Snail expressions were evaluated qualitatively (presence or absence), in nuclear and cytoplasmic cell compartments and compared in relation to clinical parameters. The Kaplan-Meier survival analysis showed the impact of the sarcomatoid component and Slug expression on the survival longevity. Cox regression analysis separated Slug as the only independent prognostic factor (p = 0.046). The expression of Snail was associated with higher stages of the disease (p = 0.004), especially observing nuclear Snail expression (p < 0.001). All of the tumors that had metastasized showed nuclear immunoreactivity (p < 0.001). In clear cell RCC, we showed a significant relationship between a high nuclear grade and nuclear Snail expression (p = 0.039). Our results suggest that Slug and Snail could be useful immunohistochemical markers for staging and prognosis in patients suffering from various RCCs, representing potential targets for further therapy strategies of renal cancer.
Collapse
Affiliation(s)
- Maja Zivotic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Sanjin Kovacevic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 9 Dr. Subotic Street, 11000 Belgrade, Serbia;
| | - Gorana Nikolic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Ana Mioljevic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Isidora Filipovic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Marija Djordjevic
- Faculty of Organization Sciences, University of Belgrade, 11010 Belgrade, Serbia;
| | - Vladimir Jovicic
- Clinic for Cardiac Surgery, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Nikola Topalovic
- Department of Medical Physiology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Kristina Ilic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Sanja Radojevic Skodric
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Dusko Dundjerovic
- Institute of Pathology, Faculty of Medicine, University of Belgrade, 1 Dr. Subotic Street, 11000 Belgrade, Serbia; (M.Z.); (G.N.); (A.M.); (I.F.); (K.I.); (S.R.S.)
| | - Jelena Nesovic Ostojic
- Department of Pathological Physiology, Faculty of Medicine, University of Belgrade, 9 Dr. Subotic Street, 11000 Belgrade, Serbia;
| |
Collapse
|
8
|
Fathi D, Elballal MS, Elesawy AE, Abulsoud AI, Elshafei A, Elsakka EG, Ismail A, El-Mahdy HA, Elrebehy MA, Doghish AS. An emphasis on the interaction of signaling pathways highlights the role of miRNAs in the etiology and treatment resistance of gastric cancer. Life Sci 2023; 322:121667. [PMID: 37023952 DOI: 10.1016/j.lfs.2023.121667] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023]
Abstract
Gastric cancer (GC) is 4th in incidence and mortality rates globally. Several genetic and epigenetic factors, including microRNAs (miRNAs), affect its initiation and progression. miRNAs are short chains of nucleic acids that can regulate several cellular processes by controlling their gene expression. So, dysregulation of miRNAs expressions is associated with GC initiation, progression, invasion capacity, apoptosis evasions, angiogenesis, promotion and EMT enhancement. Of important pathways in GC and controlled by miRNAs are Wnt/β-catenin signaling, HMGA2/mTOR/P-gp, PI3K/AKT/c-Myc, VEGFR and TGFb signaling. Hence, this review was conducted to review an updated view of the role of miRNAs in GC pathogenesis and their modulatory effects on responses to different GC treatment modalities.
Collapse
|
9
|
Zhang HF, Liu HM, Xiang JY, Zhou XC, Wang D, Chen RY, Tan WL, Liang LQ, Liu LL, Shi MJ, Zhang F, Xiao Y, Zhou YX, Zhang T, Tang L, Guo B, Wang YY. Alpha lipoamide inhibits diabetic kidney fibrosis via improving mitochondrial function and regulating RXRα expression and activation. Acta Pharmacol Sin 2023; 44:1051-1065. [PMID: 36347997 PMCID: PMC10104876 DOI: 10.1038/s41401-022-00997-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022]
Abstract
Previous studies have shown mitochondrial dysfunction in various acute kidney injuries and chronic kidney diseases. Lipoic acid exerts potent effects on oxidant stress and modulation of mitochondrial function in damaged organ. In this study we investigated whether alpha lipoamide (ALM), a derivative of lipoic acid, exerted a renal protective effect in a type 2 diabetes mellitus mouse model. 9-week-old db/db mice were treated with ALM (50 mg·kg-1·d-1, i.g) for 8 weeks. We showed that ALM administration did not affect blood glucose levels in db/db mice, but restored renal function and significantly improved fibrosis of kidneys. We demonstrated that ALM administration significantly ameliorated mitochondrial dysfunction and tubulointerstitial fibrotic lesions, along with increased expression of CDX2 and CFTR and decreased expression of β-catenin and Snail in kidneys of db/db mice. Similar protective effects were observed in rat renal tubular epithelial cell line NRK-52E cultured in high-glucose medium following treatment with ALM (200 μM). The protective mechanisms of ALM in diabetic kidney disease (DKD) were further explored: Autodock Vina software predicted that ALM could activate RXRα protein by forming stable hydrogen bonds. PROMO Database predicted that RXRα could bind the promoter sequences of CDX2 gene. Knockdown of RXRα expression in NRK-52E cells under normal glucose condition suppressed CDX2 expression and promoted phenotypic changes in renal tubular epithelial cells. However, RXRα overexpression increased CDX2 expression which in turn inhibited high glucose-mediated renal tubular epithelial cell injury. Therefore, we reveal the protective effect of ALM on DKD and its possible potential targets: ALM ameliorates mitochondrial dysfunction and regulates the CDX2/CFTR/β-catenin signaling axis through upregulation and activation of RXRα. Schematic figure illustrating that ALM alleviates diabetic kidney disease by improving mitochondrial function and upregulation and activation of RXRα, which in turn upregulated CDX2 to exert an inhibitory effect on β-catenin activation and nuclear translocation. RTEC renal tubular epithelial cell. ROS Reactive oxygen species. RXRα Retinoid X receptor-α. Mfn1 Mitofusin 1. Drp1 dynamic-related protein 1. MDA malondialdehyde. 4-HNE 4-hydroxynonenal. T-SOD Total-superoxide dismutase. CDX2 Caudal-type homeobox transcription factor 2. CFTR Cystic fibrosis transmembrane conductance regulator. EMT epithelial mesenchymal transition. α-SMA Alpha-smooth muscle actin. ECM extracellular matrix. DKD diabetic kidney disease. Schematic figure was drawn by Figdraw ( www.figdraw.com ).
Collapse
Affiliation(s)
- Hui-Fang Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Hui-Ming Liu
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jia-Yi Xiang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Xing-Cheng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Dan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Rong-Yu Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Wan-Lin Tan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Lu-Qun Liang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Ling-Ling Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Ming-Jun Shi
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Fan Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Ying Xiao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Yu-Xia Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Tian Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China
| | - Lei Tang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China.
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, 550025, China.
| | - Bing Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550025, China.
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China.
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China.
| | - Yuan-Yuan Wang
- International Scientific and Technological Cooperation Base of Pathogenesis and Drug Research on Common Major Diseases, Guizhou Medical University, Guiyang, 550025, China.
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China.
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
10
|
Qing L, Li Q, Dong Z. MUC1: An emerging target in cancer treatment and diagnosis. Bull Cancer 2022; 109:1202-1216. [DOI: 10.1016/j.bulcan.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/26/2022] [Accepted: 08/01/2022] [Indexed: 10/14/2022]
|
11
|
Chen XY, Qin XH, Xie XL, Liao CX, Liu DT, Li GW. Overexpression miR-520a-3p inhibits acute myeloid leukemia progression via targeting MUC1. Transl Oncol 2022; 22:101432. [PMID: 35649317 PMCID: PMC9156816 DOI: 10.1016/j.tranon.2022.101432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/14/2022] [Accepted: 04/11/2022] [Indexed: 11/09/2022] Open
Abstract
miR-520a-3p is downregulated and MUC1 is upregulated in AML patients. miR-520a-3p over-expression inhibits AML cell proliferation, accelerates apoptosis, and inhibits AML tumor growth. miR-520a-3p targets MUC1 and negatively regulates its expression. MUC1 knockdown supresses AML cell proliferation and promotes apoptosis. miR-520a-3p overexpression inhibits AML cell proliferation and accelerates cell apoptosis via regulating MUC1/Wnt/β-catenin axis.
Background Acute myeloid leukemia (AML) is one of the familiar malignant tumors in the hematological system. miR-520a-3p is reported to be involved in several cancers’ progression. However, miR-520a-3p role in AML remains unclear. In this study, we aimed to clarify the role and potential mechanism of miR-520a-3p in AML. Methods Cell viability, proliferation, cycle and apoptosis were detected by MTT assay, colony formation assay, flow cytometry, respectively. The levels of PNCA, Bcl-2, Cleaved caspase 3, Cleaved caspase 9 and β-catenin protein were detected by Western blot. Dual-luciferase reported assay was performed to detect the regulation between miR-520a-3p and MUC1. To verify the effect of miR-520a-3p on tumor proliferation in vivo, a non-homogenous transplant model of tumors was established. Results miR-520a-3p expression was down-regulated, and MUC1 expression was up-regulated in AML patients. miR-520a-3p overexpression suppressed THP-1 cell proliferation, induced cell cycle G0/G1 inhibition and promoted apoptosis. miR-520a-3p targeted MUC1 and negatively regulated its expression. MUC1 knockdown inhibited THP-1 cell proliferation and promoted apoptosis. miR-520a-3p overexpression inhibited AML tumors growth. Conclusion Overexpression miR-520a-3p inhibited AML cell proliferation, and promoted apoptosis via inhibiting MUC1 expression and repressing Wnt/β-catenin pathway activation.
Collapse
|
12
|
Kufe DW. Chronic activation of MUC1-C in wound repair promotes progression to cancer stem cells. JOURNAL OF CANCER METASTASIS AND TREATMENT 2022; 8. [PMID: 35539431 PMCID: PMC9083497 DOI: 10.20517/2394-4722.2022.03] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mucin 1 (MUC1) gene emerged in mammals to afford protection of barrier epithelial tissues from the external environment. MUC1 encodes a transmembrane C-terminal (MUC1-C) subunit that is activated by loss of homeostasis and induces inflammatory, proliferative, and remodeling pathways associated with wound repair. As a consequence, chronic activation of MUC1-C promotes lineage plasticity, epigenetic reprogramming, and carcinogenesis. In driving cancer progression, MUC1-C is imported into the nucleus, where it induces NF-κB inflammatory signaling and the epithelial-mesenchymal transition (EMT). MUC1-C represses gene expression by activating (i) DNA methyltransferase 1 (DNMT1) and DNMT3b, (ii) Polycomb Repressive Complex 1 (PRC1) and PRC2, and (iii) the nucleosome remodeling and deacetylase (NuRD) complex. PRC1/2-mediated gene repression is counteracted by the SWI/SNF chromatin remodeling complexes. MUC1-C activates the SWI/SNF BAF and PBAF complexes in cancer stem cell (CSC) models with the induction of genome-wide differentially accessible regions and expressed genes. MUC1-C regulates chromatin accessibility of enhancer-like signatures in association with the induction of the Yamanaka pluripotency factors and recruitment of JUN and BAF, which promote increases in histone activation marks and opening of chromatin. These and other findings described in this review have uncovered a pivotal role for MUC1-C in integrating lineage plasticity and epigenetic reprogramming, which are transient in wound repair and sustained in promoting CSC progression.
Collapse
Affiliation(s)
- Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Hosseinzadeh A, Merikhian P, Naseri N, Eisavand MR, Farahmand L. MUC1 is a potential target to overcome trastuzumab resistance in breast cancer therapy. Cancer Cell Int 2022; 22:110. [PMID: 35248049 PMCID: PMC8897942 DOI: 10.1186/s12935-022-02523-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/12/2022] [Indexed: 02/07/2023] Open
Abstract
Although resistance is its major obstacle in cancer therapy, trastuzumab is the most successful agent in treating epidermal growth factor receptor 2 positive (HER2 +) breast cancer (BC). Some patients show resistance to trastuzumab, and scientists want to circumvent this problem. This review elaborately discusses possible resistance mechanisms to trastuzumab and introduces mucin 1 (MUC1) as a potential target efficient for overcoming such resistance. MUC1 belongs to the mucin family, playing the oncogenic/mitogenic roles in cancer cells and interacting with several other oncogenic receptors and pathways, such as HER2, β-catenin, NF-κB, and estrogen receptor (ERα). Besides, it has been established that MUC1- Cytoplasmic Domain (MUC1-CD) accelerates the development of resistance to trastuzumab and that silencing MUC1-C proto-oncogene is associated with increased sensitivity of HER2+ cells to trastuzumab-induced growth inhibitors. We mention why targeting MUC1 can be useful in overcoming trastuzumab resistance in cancer therapy.
Collapse
|
14
|
Goujon M, Woszczyk J, Gaudelot K, Swierczewski T, Fellah S, Gibier JB, Van Seuningen I, Larrue R, Cauffiez C, Gnemmi V, Aubert S, Pottier N, Perrais M. A Double-Negative Feedback Interaction between miR-21 and PPAR-α in Clear Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14030795. [PMID: 35159062 PMCID: PMC8834244 DOI: 10.3390/cancers14030795] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/04/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the main histotype of kidney cancer, which is typically highly resistant to conventional therapies and known for abnormal lipid accumulation. In this context, we focused our attention on miR-21, an oncogenic miRNA overexpressed in ccRCC, and peroxysome proliferator-activated receptor-α (PPAR- α), one master regulator of lipid metabolism targeted by miR-21. First, in a cohort of 52 primary ccRCC samples, using RT-qPCR and immunohistochemistry, we showed that miR-21 overexpression was correlated with PPAR-α downregulation. Then, in ACHN and 786-O cells, using RT-qPCR, the luciferase reporter gene, chromatin immunoprecipitation, and Western blotting, we showed that PPAR-α overexpression (i) decreased miR-21 expression, AP-1 and NF-κB transcriptional activity, and the binding of AP-1 and NF-κB to the miR-21 promoter and (ii) increased PTEN and PDCD4 expressions. In contrast, using pre-miR-21 transfection, miR-21 overexpression decreased PPAR-α expression and transcriptional activity mediated by PPAR-α, whereas the anti-miR-21 (LNA-21) strategy increased PPAR-α expression, but also the expression of its targets involved in fatty acid oxidation. In this study, we showed a double-negative feedback interaction between miR-21 and PPAR-α. In ccRCC, miR-21 silencing could be therapeutically exploited to restore PPAR-α expression and consequently inhibit the oncogenic events mediated by the aberrant lipid metabolism of ccRCC.
Collapse
Affiliation(s)
- Marine Goujon
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Justine Woszczyk
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Kelly Gaudelot
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Thomas Swierczewski
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Sandy Fellah
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Jean-Baptiste Gibier
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- CHU Lille, Service d’Anatomo-Pathologie, F-59000 Lille, France
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Romain Larrue
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- CHU Lille, Service de Toxicologie et Génopathies, F-59000 Lille, France
| | - Christelle Cauffiez
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
| | - Viviane Gnemmi
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- CHU Lille, Service d’Anatomo-Pathologie, F-59000 Lille, France
| | - Sébastien Aubert
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- CHU Lille, Service d’Anatomo-Pathologie, F-59000 Lille, France
| | - Nicolas Pottier
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- CHU Lille, Service de Toxicologie et Génopathies, F-59000 Lille, France
| | - Michaël Perrais
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.G.); (J.W.); (K.G.); (T.S.); (S.F.); (J.-B.G.); (I.V.S.); (R.L.); (C.C.); (V.G.); (S.A.); (N.P.)
- Correspondence: ; Tel.: +33-3-20-29-88-62
| |
Collapse
|
15
|
Gao W, Zhang S, Guorong L, Liu Q, Zhu A, Gui F, Zou Y, Wu Y, Luo Y, Hong Z. Nc886 promotes renal cancer cell drug-resistance by enhancing EMT through Rock2 phosphorylation-mediated β-catenin nuclear translocation. Cell Cycle 2022; 21:340-351. [PMID: 34974812 PMCID: PMC8855853 DOI: 10.1080/15384101.2021.2020431] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Drug resistance is a significant challenge in the present treatment regimens of renal cell carcinoma (RCC). Our previous study confirmed that nc886 functions as an oncogene in RCC. Nevertheless, the role and underlying mechanism of nc886 in RCC drug resistance are unclear. In the present study, Sunitinib and Everolimus treatment, respectively, downregulated nc886 expression in a dose-dependent manner in all four renal cancer cell lines. Nc886 overexpression in 786-O cells and ACHN cells significantly reduced the sensitivity of cancer cells to both Sunitinib and Everolimus treatment, respectively, by promoting cell viability and inhibiting cell apoptosis, whereas nc886 silencing increased cancer cell sensitivity. In renal cancer cell line with the highest drug-resistance, 786-O cells, Sunitinib, or Everolimus treatment enhanced the cellular EMT and was further enhanced by nc886 overexpression while attenuated by nc886 silencing. In 786-O cells, nc886 overexpression significantly promoted EMT, ROCK2 phosphorylation, and β-catenin nucleus translocation under Sunitinib or Everolimus treatment. Moreover, ROCK2 silencing significantly reversed the effects of nc886 overexpression on EMT, ROCK2 phosphorylation, and β-catenin nucleus translocation, as well as drug-resistant renal cancer cell viability and apoptosis. In conclusion, it was demonstrated that nc886 promotes renal cancer cell proliferation, migration, and invasion, as demonstrated previously. nc886 also promotes renal cancer cell drug-resistance to Sunitinib or Everolimus by promoting EMT through Rock2 phosphorylation-mediated nuclear translocation of β-catenin.
Collapse
Affiliation(s)
- Weiyin Gao
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Children’s Hospital, Nanchang, Jiangxi Province, China
| | - Li Guorong
- Department of Urology, North Hospital, Chu Saint-Etienne, University of Jean-Monnet, Saint-Etienne, France
| | - Queling Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Anyi Zhu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Fu Gui
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yan Zou
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yiguo Wu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yang Luo
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhengdong Hong
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China,CONTACT Zhengdong Hong The Second Affiliated Hospital of Nanchang University, No.1 Minde Road, Nanchang, Jiangxi Province, China
| |
Collapse
|
16
|
Inhibition of FSTL3 abates the proliferation and metastasis of renal cell carcinoma via the GSK-3β/β-catenin signaling pathway. Aging (Albany NY) 2021; 13:22528-22543. [PMID: 34555811 PMCID: PMC8507290 DOI: 10.18632/aging.203564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is a lethal malignancy of the genitourinary system. Follistatin-like 3 (FSTL3), which mediates cell differentiation and growth, acts as a biomarker of tumors and participates in cancer development and progression. Presently, the FSTL3’s functions in RCC were investigated. Quantitative reverse transcription PCR (qRT-PCR), Western Blot, and enzyme linked immunosorbent assay (ELISA) were conducted to verify FSTL3 expression in RCC tissues and cell lines. BrdU assay and CCK8 experiment were made to monitor cell proliferation. Transwell was implemented to examine the invasion of the cells. Flow cytometry analyzed cell apoptosis, and Western Blot evaluated the protein levels of E-cadherin, Twist, and Slug. In the meantime, the protein profiles of the GSK-3β, β-catenin, and TGF-β signaling pathways were ascertained. Moreover, the Xenograft tumor model was constructed in nude mice for measuring tumor growth in vivo. The statistics showed that FSTL3 presented an overexpression in RCC, and patients with a lower expression of FSTL3 manifested a better prognosis. Down-regulated FSTL3 hampered the proliferation, invasion, EMT, and tumor growth of RCC cells and caused cell apoptosis. On the contrary, FSTL3 overexpression enhanced the malignant behaviors of RCC cells. Furthermore, FSTL3 knockdown bolstered GSK-3β, suppressed β-catenin, and reduced BMP1-SMAD pathway activation. Inhibited β-catenin substantially mitigated FSTL3-mediated promoting functions in RCC. In short, FSTL3 functions as an oncogene in RCC by modulating the GSK-3β/β-catenin signaling pathway.
Collapse
|
17
|
Tao Y, Han J, Liu W, An L, Hu W, Wang N, Yu Y. MUC1 Promotes Mesangial Cell Proliferation and Kidney Fibrosis in Diabetic Nephropathy Through Activating STAT and β-Catenin Signal Pathway. DNA Cell Biol 2021; 40:1308-1316. [PMID: 34520253 DOI: 10.1089/dna.2021.0098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes, which leads to most end-stage kidney diseases and threatens health of patients. Mucin 1 (MUC1) is a heterodimeric oncoprotein, which is abnormally expressed in tumors and hematologic diseases. The aim of this study is to clarify the mechanism and role of MUC1 in DN. The mesangial cells (MCs) suffered from high glucose (HG) treatment to mimic DN in vitro. The cell proliferation was detected by Cell Counting Kit-8 assay and 5-ethynyl-2-deoxyuridine (EdU) staining assay. The expression of MUC1 and fibrosis markers: fibronectin, collagen I, and collagen IV were assessed by western blot. In this study, we demonstrated that HG treatment induced MUC1 expression in MCs. With knockdown of MUC1 or overexpressed MUC1 in MCs, the results indicated that knockdown of MUC1 inhibited MCs proliferation and reduced kidney fibrosis markers expression, including fibronectin, collagen I, and collagen IV, whereas overexpression of MUC1 led to opposite results. Mechanically, MUC1 activated signal transducers and activators of transcription (STAT) and β-catenin signal pathway. After added AG490 (STAT inhibitor) or FH535 (β-catenin inhibitor), blocking STAT3 and β-catenin signal pathway attenuated MUC1-induced cell proliferation and fibronectin production in MCs. Finally, knockdown of MUC1 attenuated DN-induced kidney fibrosis in db/db mice. Therapeutic target for DN. In conclusion, MUC1 promotes MCs proliferation and kidney fibrosis in DN through activating STAT and β-catenin signal pathway, which can help to provide a novel therapeutic target for DN.
Collapse
Affiliation(s)
- Yiying Tao
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Jianfang Han
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Wenhua Liu
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Ling An
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Wenbo Hu
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Ningning Wang
- Department of Nephrology, Qinghai Provincial People's Hospital, Xining City, China
| | - Yean Yu
- Department of Nephrology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan City, China
| |
Collapse
|
18
|
Zubareva EY, Senchukova MA. Prognostic and predictive significance of osteopontin in malignant neoplasms. ADVANCES IN MOLECULAR ONCOLOGY 2021; 8:23-28. [DOI: 10.17650/2313-805x-2021-8-2-23-28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Osteopontin is an extracellular matrix protein which is produced by different types of cells and plays an important functional role in many biological processes. This review discusses the main functions of osteopontin, its role in the progression and chemoresistance of malignant neoplasms, in the regulation of epithelial-mesenchymal transition, angiogenesis, and the body’s immune response to the tumor. The article considers the currently known mechanisms by which osteopontin affects to the survival, mobility and invasion of tumor cells, to tumor sensitivity to drug treatment, as well as the prospects for a integrated study of the predictive significance of osteopontin, markers of hypoxia, angiogenesis, epithelial- mesenchymal transition, and immunological tolerance.
Collapse
Affiliation(s)
- E. Yu. Zubareva
- Orenburg Regional Clinical Oncological Dispensary; Orenburg State Medical University
| | - M. A. Senchukova
- Orenburg Regional Clinical Oncological Dispensary; Orenburg State Medical University
| |
Collapse
|
19
|
Shi Y, Xu Y, Yao J, Yan C, Su H, Zhang X, Chen E, Ying K. MTHFD2 promotes tumorigenesis and metastasis in lung adenocarcinoma by regulating AKT/GSK-3β/β-catenin signalling. J Cell Mol Med 2021; 25:7013-7027. [PMID: 34121323 PMCID: PMC8278097 DOI: 10.1111/jcmm.16715] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 01/11/2023] Open
Abstract
Recent studies have demonstrated that one‐carbon metabolism plays a significant role in cancer development. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a mitochondrial enzyme of one‐carbon metabolism, has been reported to be dysregulated in many cancers. However, the specific role and mechanism of MTHFD2 in lung adenocarcinoma (LUAD) still remains unclear. In this study, we evaluated the clinicopathological and prognostic values of MTHFD2 in LUAD patients. We conducted a series of functional experiments in vivo and in vitro to explore novel mechanism of MTHFD2 in LUAD. The results showed that MTHFD2 was significantly up‐regulated in LUAD tissues and predicted poor prognosis of LUAD patients. Knockdown of MTHFD2 dramatically inhibited cell proliferation and migration by blocking the cell cycle and inducing the epithelial‐mesenchymal transition (EMT). In addition, MTHFD2 knockdown suppressed LUAD growth and metastasis in cell‐derived xenografts. Mechanically, we found that MTHFD2 promoted LUAD cell growth and metastasis via AKT/GSK‐3β/β‐catenin signalling. Finally, we identified miR‐30a‐3p as a novel regulator of MTHFD2 in LUAD. Collectively, MTHFD2 plays an oncogenic role in LUAD progression and is a promising target for LUAD diagnosis and therapy.
Collapse
Affiliation(s)
- Yangfeng Shi
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Yiming Xu
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Jianchang Yao
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Chao Yan
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Hua Su
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Enguo Chen
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Kejing Ying
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Kufe DW. MUC1-C in chronic inflammation and carcinogenesis; emergence as a target for cancer treatment. Carcinogenesis 2021; 41:1173-1183. [PMID: 32710608 DOI: 10.1093/carcin/bgaa082] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is a highly prevalent consequence of changes in environmental and lifestyle factors that contribute to the development of cancer. The basis for this critical association has largely remained unclear. The MUC1 gene evolved in mammals to protect epithelia from the external environment. The MUC1-C subunit promotes responses found in wound healing and cancer. MUC1-C induces EMT, epigenetic reprogramming, dedifferentiation and pluripotency factor expression, which when prolonged in chronic inflammation promote cancer progression. As discussed in this review, MUC1-C also drives drug resistance and immune evasion, and is an important target for cancer therapeutics now under development.
Collapse
Affiliation(s)
- Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Huffstater T, Merryman WD, Gewin LS. Wnt/β-Catenin in Acute Kidney Injury and Progression to Chronic Kidney Disease. Semin Nephrol 2021; 40:126-137. [PMID: 32303276 DOI: 10.1016/j.semnephrol.2020.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) portends a poor clinical prognosis and increases the risk for the development of chronic kidney disease (CKD). Currently, there are no therapies to treat AKI or prevent its progression to CKD. Wnt/β-catenin is a critical regulator of kidney development that is up-regulated after injury. Most of the literature support a beneficial role for Wnt/β-catenin in AKI, but suggest that this pathway promotes the progression of tubulointerstitial fibrosis, the hallmark of CKD progression. We review the role of Wnt/β-catenin in renal injury with a focus on its potential as a therapeutic target in AKI and in AKI to CKD transition.
Collapse
Affiliation(s)
- Tessa Huffstater
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Leslie S Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Medicine, Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, TN; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN.
| |
Collapse
|
22
|
Menju T, Date H. Lung cancer and epithelial-mesenchymal transition. Gen Thorac Cardiovasc Surg 2021; 69:781-789. [PMID: 33754237 DOI: 10.1007/s11748-021-01595-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer (LC) is a leading cause of cancer-related death worldwide. Epithelial-mesenchymal transition (EMT) is a well-known phenomenon that promotes the invasive and metastatic capabilities of LC. Especially, EMT is assumed to be a pivotal mechanism for tumor cell invasion and metastasis, thereby limiting the efficacy of surgery and medical treatments, resulting in poor patient prognoses. Thus, the elucidation and reversal of EMT could provide changes in therapeutic strategies for LC. To overcome the limitations of currents treatment regimens for LC, it is important for surgeons to be familiar with this complex tumor characteristic. In this review, the activating signaling pathways underlying EMT and the associated tumor phenotypes are briefly described.
Collapse
Affiliation(s)
- Toshi Menju
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54, Shogoin Kawara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
23
|
Liu H, Yan R, Liang L, Zhang H, Xiang J, Liu L, Zhang X, Mao Y, Peng W, Xiao Y, Zhang F, Zhou Y, Shi M, Wang Y, Guo B. The role of CDX2 in renal tubular lesions during diabetic kidney disease. Aging (Albany NY) 2021; 13:6782-6803. [PMID: 33621200 PMCID: PMC7993706 DOI: 10.18632/aging.202537] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/09/2020] [Indexed: 02/05/2023]
Abstract
Renal tubules are vulnerable targets of various factors causing kidney injury in diabetic kidney disease (DKD), and the degree of tubular lesions is closely related to renal function. Abnormal renal tubular epithelial cells (RTECs) differentiation and depletion of cell junction proteins are important in DKD pathogenesis. Caudal-type homeobox transcription factor 2 (CDX2), represents a key nuclear transcription factor that maintains normal proliferation and differentiation of the intestinal epithelium. The present study aimed to evaluate the effects of CDX2 on RTECs differentiation and cell junction proteins in DKD. The results demonstrated that CDX2 was mainly localized in renal tubules, and downregulated in various DKD models. CDX2 upregulated E-cadherin and suppressed partial epithelial-mesenchymal transition (EMT), which can alleviate hyperglycemia-associated RTECs injury. Cystic fibrosis transmembrane conductance regulator (CFTR) was regulated by CDX2 in NRK-52E cells, and CFTR interfered with β-catenin activation by binding to Dvl2, which is an essential component of Wnt/β-catenin signaling. CFTR knockdown abolished the suppressive effects of CDX2 on Wnt/β-catenin signaling, thereby upregulating cell junction proteins and inhibiting partial EMT in RTECs. In summary, CDX2 can improve renal tubular lesions during DKD by increasing CFTR amounts to suppress the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Huiming Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Luqun Liang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Huifang Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Jiayi Xiang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Lingling Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Xiaohuan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yanwen Mao
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Wei Peng
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang 550025, Guizhou, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang 550025, Guizhou, China
| |
Collapse
|
24
|
MUC1 Mitigates Renal Injury and Inflammation in Endotoxin Induced Acute Kidney Injury by Inhibiting the TLR4-MD2 Axis and Reducing Pro-Inflammatory Macrophages Infiltration. Shock 2021; 56:629-638. [PMID: 33534395 DOI: 10.1097/shk.0000000000001742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Sepsis is the leading cause of acute kidney injury (AKI) in critical care patients. A cornerstone of sepsis-associated AKI is dysregulated inflammation driven by excessive activation of Toll-like receptor 4 (TLR4) pathway. MUC1, a membrane bound mucin expressed in both epithelial tubular cells and renal macrophages, has been shown to be involved in the regulation of TLRs. Therefore we hypothesized that MUC1 could mitigate the renal inflammatory response to TLR4 activation. To test this hypothesis, we used a murine model of endotoxin-induced AKI by intraperitoneal injection of lipopolysaccharide (LPS). We showed that Muc1-/- mice have a more severe renal dysfunction, an increased activation of the tissular NF-kB pathway and secreted more pro inflammatory cytokines compare to Muc1+/+ mice. By flow cytometry, we observed that the proportion of M1 (pro-inflammatory) macrophages in the kidneys of Muc1-/- mice was significantly increased. In human and murine primary macrophages, we showed that MUC1 is only induced in M1 type macrophages and that macrophages derived from Muc1-/- mice secreted more pro-inflammatory cytokines. Eventually, in HEK293 cells, we showed that (i) MUC1 cytosolic domain (CT) seems necessary for the negative regulation of TLR4 (ii) by proximity ligation assay, MUC1-CT is in close relationship with TLR4 and acts as a competitive inhibitor of the recruitment of MYD88. Overall our results support that in the context of endotoxin-induced AKI, MUC1 plays a significant role in controlling disease severity by regulating negatively the TLR4-MD2 axis.
Collapse
|
25
|
Bali P, Lozano-Pope I, Pachow C, Obonyo M. Early detection of tumor cells in bone marrow and peripheral blood in a fast‑progressing gastric cancer model. Int J Oncol 2021; 58:388-396. [PMID: 33469673 PMCID: PMC7864146 DOI: 10.3892/ijo.2021.5171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection is a major risk factor for the development of gastric cancer. The authors previously demonstrated that in mice deficient in myeloid differentiation primary response 88 (Myd88−/−), infection with Helicobacter felis (H. felis) a close relative of H. pylori, subsequently rapidly progressed to neoplasia. The present study examined circulating tumor cells (CTCs) by measuring the expression of cytokeratins, epithelial-to-mesenchymal transition (EMT)-related markers and cancer stem cell (CSC) markers in bone marrow and peripheral blood from Myd88−/− and wild-type (WT) mice. Cytokeratins CK8/18 were detected as early as 4 months post-infection in Myd88−/− mice. By contrast, cytokeratins were not detected in WT mice even after 7 months post-infection. The expression of Mucin-1 (MUC1) was observed in both bone marrow and peripheral blood at different time points, suggesting its role in gastric cancer metastasis. Snail, Twist and ZEB were expressed at different levels in bone marrow and peripheral blood. The expression of these EMT-related markers suggests the manifestation of cancer metastasis in the early stages of disease development. LGR5, CD44 and CD133 were the most prominent CSC markers detected. The detection of CSC and EMT markers along with cytokeratins does reinforce their use as biomarkers for gastric cancer metastasis. This early detection of markers suggests that CTCs leave primary site even before cancer is well established. Thus, cytokeratins, EMT, and CSCs could be used as biomarkers to detect aggressive forms of gastric cancers. This information may prove to be of significance in stratifying patients for treatment prior to the onset of severe disease-related characteristics.
Collapse
Affiliation(s)
- Prerna Bali
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Ivonne Lozano-Pope
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Collin Pachow
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| | - Marygorret Obonyo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093‑0640, USA
| |
Collapse
|
26
|
GATA6 promotes epithelial-mesenchymal transition and metastasis through MUC1/β-catenin pathway in cholangiocarcinoma. Cell Death Dis 2020; 11:860. [PMID: 33060563 PMCID: PMC7567063 DOI: 10.1038/s41419-020-03070-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 01/06/2023]
Abstract
GATA6 acts as an oncogene or tumour suppressor in different cancers. Previously, we found that aberrant expression of GATA6 promoted metastasis in cholangiocarcinoma (CCA). However, the mechanism by which GATA6 promotes metastasis in CCA is unclear. In the present study, we aimed to investigate the role of GATA6 in CCA cell epithelial–mesenchymal transition (EMT). Our results showed that GATA6 expression was positively associated with N-cadherin and vimentin expression but negatively associated with E-cadherin expression in 91 CCA samples. GATA6 promoted EMT and metastasis in CCA cells in vitro and in vivo based on knockdown and overexpression analyses. ChIP-sequencing data revealed that MUC1 is a novel downstream target of GATA6. GATA6 upregulated MUC1 expression through binding to both the 1584 and 1456 GATA-motifs in the promoter region and enhancing its transcription by luciferase reporter assays and point-mutant assays. MUC1 expression was positively associated with N-cadherin and vimentin expression but negatively associated with E-cadherin expression in 91 CCA samples. In addition, MUC1 promoted EMT in CCA cells based on knockdown and overexpression analyses. Moreover, MUC1 knockdown significantly abrogated the GATA6-induced EMT in CCA cells, indicating that MUC1 promoted EMT through upregulating MUC1 in CCA cells. β-Catenin is a putative transcriptional coactivator that regulates EMT in cancers. Our data showed that MUC1 expression was positively associated with nuclear β-catenin expression in 91 CCA samples. MUC1 upregulated nuclear β-catenin expression in CCA cells. Moreover, MUC1 bound to β-catenin in CCA cells based on protein immunoprecipitation analyses. MUC1 knockdown significantly decreased the binding of MUC1 to β-catenin, and thereby decreased nuclear β-catenin protein levels in CCA cells, indicating that MUC1 bound to β-catenin and increased its nuclear expression in CCA cells. Together, our results show that GATA6 promotes EMT through MUC1/β-catenin pathway in CCA, indicating potential implications for anti-metastatic therapy.
Collapse
|
27
|
Sohn SH, Sul HJ, Kim B, Kim BJ, Kim HS, Zang DY. Tepotinib Inhibits the Epithelial-Mesenchymal Transition and Tumor Growth of Gastric Cancers by Increasing GSK3β, E-Cadherin, and Mucin 5AC and 6 Levels. Int J Mol Sci 2020; 21:ijms21176027. [PMID: 32825724 PMCID: PMC7503648 DOI: 10.3390/ijms21176027] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022] Open
Abstract
Aberrant expression of mucins (MUCs) can promote the epithelial–mesenchymal transition (EMT), which leads to enhanced tumorigenesis. Carcinogenesis-related pathways involving c-MET and β-catenin are associated with MUCs. In this study, we characterized the expression of EMT-relevant proteins including MET, β-catenin, and E-cadherin in human gastric cancer (GC) cell lines, and further characterized the differential susceptibility of these cell lines compared with the c-MET inhibitor tepotinib. We assessed the antitumor activity of tepotinib in GC cell lines. The effects of tepotinib on cell viability, apoptotic cell death, EMT, and c-MET and β-catenin signaling were evaluated by 3-(4,5 dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl-2-(4-sulfophenyl)-2H-tetrazolium (MTS), flow cytometry, Western blotting, and qRT-PCR. The antitumor efficacy was assessed in MKN45 xenograft mice. Tepotinib treatment induced apoptosis in c-MET-amplified SNU620, MKN45, and KATO III cells, but had no effect on c-MET-reduced MKN28 or AGS cells. Tepotinib treatment also significantly reduced the protein levels of phosphorylated and total c-MET, phosphorylated and total ERK, β-catenin, and c-MYC in SNU620 and MKN45 cells. In contrast, this drug was only slightly active against KATO III cells. Notably, tepotinib significantly reduced the expression of EMT-promoting genes such as MMP7, COX-2, WNT1, MUC5B, and c-MYC in c-MET-amplified GC cells and increased the expression of EMT-suppressing genes such as MUC5AC, MUC6, GSK3β, and E-cadherin. In a mouse model, tepotinib exhibited good antitumor growth activity along with increased E-cadherin and decreased phosphorylated c-MET (phospho-c-MET) protein levels. Collectively, these results suggest that tepotinib suppresses tumor growth and migration by negatively regulating c-MET-induced EMT. These findings provide new insights into the mechanism by which MUC5AC and MUC6 contribute to GC progression.
Collapse
Affiliation(s)
- Sung-Hwa Sohn
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang 14066, Korea; (S.-H.S.); (H.J.S.); (B.K.)
| | - Hee Jung Sul
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang 14066, Korea; (S.-H.S.); (H.J.S.); (B.K.)
| | - Bohyun Kim
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang 14066, Korea; (S.-H.S.); (H.J.S.); (B.K.)
| | - Bum Jun Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Korea; (B.J.K.); (H.S.K.)
| | - Hyeong Su Kim
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Korea; (B.J.K.); (H.S.K.)
| | - Dae Young Zang
- Hallym Translational Research Institute, Hallym University Sacred Heart Hospital, Anyang 14066, Korea; (S.-H.S.); (H.J.S.); (B.K.)
- Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Anyang-si, Gyeonggi-do 14068, Korea; (B.J.K.); (H.S.K.)
- Correspondence: ; Tel.: +82-31-380-4167
| |
Collapse
|
28
|
Reynolds IS, Fichtner M, McNamara DA, Kay EW, Prehn JHM, Burke JP. Mucin glycoproteins block apoptosis; promote invasion, proliferation, and migration; and cause chemoresistance through diverse pathways in epithelial cancers. Cancer Metastasis Rev 2020; 38:237-257. [PMID: 30680581 DOI: 10.1007/s10555-019-09781-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overexpression of mucin glycoproteins has been demonstrated in many epithelial-derived cancers. The significance of this overexpression remains uncertain. The aim of this paper was to define the association of mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers by performing a systematic review of all published data. A systematic review of PubMed, Embase, and the Cochrane Central Register of Controlled Trials was performed to identify all papers that evaluated the association between mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers. PRISMA guidelines were adhered to. Results of individual studies were extracted and pooled together based on the organ in which the cancer was derived from. The initial search revealed 2031 papers, of which 90 were deemed eligible for inclusion in the study. The studies included details on MUC1, MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16. The majority of studies evaluated MUC1. MUC1 overexpression was consistently associated with resistance to apoptosis and resistance to chemotherapy. There was also evidence that overexpression of MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16 conferred resistance to apoptosis in epithelial-derived cancers. The overexpression of mucin glycoproteins is associated with resistance to apoptosis in numerous epithelial cancers. They cause resistance through diverse signaling pathways. Targeting the expression of mucin glycoproteins represents a potential therapeutic target in the treatment of epithelial-derived cancers.
Collapse
Affiliation(s)
- Ian S Reynolds
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Michael Fichtner
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Deborah A McNamara
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Elaine W Kay
- Department of Pathology, Beaumont Hospital, Dublin 9, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - John P Burke
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
29
|
Milara J, Ballester B, Montero P, Escriva J, Artigues E, Alós M, Pastor-Clerigues A, Morcillo E, Cortijo J. MUC1 intracellular bioactivation mediates lung fibrosis. Thorax 2019; 75:132-142. [PMID: 31801904 DOI: 10.1136/thoraxjnl-2018-212735] [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: 10/17/2018] [Revised: 11/05/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Serum KL6/mucin 1 (MUC1) has been identified as a potential biomarker in idiopathic pulmonary fibrosis (IPF), but the role of MUC1 intracellular bioactivation in IPF is unknown. OBJECTIVE To characterise MUC1 intracellular bioactivation in IPF. METHODS AND RESULTS The expression and phosphorylation of Thr41 and Tyr46 on the intracellular MUC1-cytoplasmic tail (CT) was increased in patients with IPF (n=22) compared with healthy subjects (n=21) and localised to fibroblasts and hyperplastic alveolar type II cells. Transforming growth factor (TGF)-β1 phosphorylated SMAD3 and thereby increased the phosphorylation of MUC1-CT Thr41 and Tyr46 in lung fibroblasts and alveolar type II cells, activating β-catenin to form a phospho-Smad3/MUC1-CT and MUC1-CT/β-catenin nuclear complex. This nuclear complex promoted alveolar epithelial type II and fibroblast to myofibroblast transitions, as well as cell senescence and fibroblast proliferation. The inhibition of MUC1-CT nuclear translocation using the inhibitor, GO-201 or silencing MUC1 by siRNA, reduced myofibroblast transition, senescence and proliferation in vitro. Bleomycin-induced lung fibrosis was reduced in mice treated with GO-201 and in MUC1-knockout mice. The profibrotic lectin, galectin-3, directly activated MUC1-CT and served as a bridge between the TGF-β receptor and the MUC1-C domain, indicating TGF-β1-dependent and TGF-β1-independent intracellular bioactivation of MUC1. CONCLUSIONS MUC1 intracellular bioactivation is enhanced in IPF and promotes fibrotic processes that could represent potential druggable targets for IPF.
Collapse
Affiliation(s)
- Javier Milara
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Beatriz Ballester
- Pharmacology, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Paula Montero
- Farmacología, Universitat de Valencia Facultat de Medicina i Odontologia, Valencia, Spain
| | - Juan Escriva
- Thoracic Surgery, Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | - Enrique Artigues
- Surgery, Hospital General Universitari de València, Valencia, Spain
| | - Manuel Alós
- Pharmacy, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Alfonso Pastor-Clerigues
- Unidad de Investigación y Docencia, Consorci Hospital General Universitari de Valencia, Valencia, Spain
| | | | - Julio Cortijo
- Pharmacology, University of Valencia, Valencia, Spain
| |
Collapse
|
30
|
Xiong W, Zhang L, Liu H, Li N, Du Y, He H, Zhang Z, Liu Y. E 2 -mediated EMT by activation of β-catenin/Snail signalling during the development of ovarian endometriosis. J Cell Mol Med 2019; 23:8035-8045. [PMID: 31560827 PMCID: PMC6850947 DOI: 10.1111/jcmm.14668] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/15/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Endometriosis is an oestrogen-dependent disease, and epithelial-mesenchymal transition (EMT) is involved in the process of endometriosis. Whether oestrogen could induce EMT in endometriosis remains largely unknown. Here, we reported that up-regulated expression of EMT markers in ovarian chocolate cyst is accompanied by high expression 17β-hydroxysteroid dehydrogenase 1 (17β-HSD1), and exposure of primary human endometrial epithelial cells to oestradiol conditions could promote EMT occurrence and activate both β-catenin and Snail signalling. Furthermore, we found nuclear β-catenin and Snail expression was closely linked in ovarian endometriosis, and β-catenin knockdown abrogated oestrogen-induced Snail mediated EMT in vitro. This is due to that β-catenin/ TCF-3 could bind to Snail promoter and activate its transcription. These results suggested that β-catenin signalling functions as the Snail activator and plays a critical role in oestradiol-induced EMT in endometriosis.
Collapse
Affiliation(s)
- Wenqian Xiong
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ling Zhang
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hengwei Liu
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Na Li
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yu Du
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Haitang He
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhibing Zhang
- Department of Obstetrics and GynecologyVirginia Commonwealth UniversityRichmondVirginia
| | - Yi Liu
- Department of Obstetrics and GynecologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
31
|
Hata T, Rajabi H, Yamamoto M, Jin C, Ahmad R, Zhang Y, Kui L, Li W, Yasumizu Y, Hong D, Miyo M, Hiraki M, Maeda T, Suzuki Y, Takahashi H, Samur M, Kufe D. Targeting MUC1-C Inhibits TWIST1 Signaling in Triple-Negative Breast Cancer. Mol Cancer Ther 2019; 18:1744-1754. [PMID: 31308076 DOI: 10.1158/1535-7163.mct-19-0156] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/21/2019] [Accepted: 07/08/2019] [Indexed: 01/06/2023]
Abstract
The oncogenic MUC1-C protein and the TWIST1 epithelial-mesenchymal transition transcription factor (EMT-TF) are aberrantly expressed in triple-negative breast cancer (TNBC) cells. However, there is no known association between MUC1-C and TWIST1 in TNBC or other cancer cells. Here, we show that MUC1-C activates STAT3, and that MUC1-C and pSTAT3 drive induction of the TWIST1 gene. In turn, MUC1-C binds directly to TWIST1, and MUC1-C/TWIST1 complexes activate MUC1-C expression in an autoinductive circuit. The functional significance of the MUC1-C/TWIST1 circuit is supported by the demonstration that this pathway is sufficient for driving (i) the EMT-TFs, ZEB1 and SNAIL, (ii) multiple genes in the EMT program as determined by RNA-seq, and (iii) the capacity for cell invasion. We also demonstrate that the MUC1-C/TWIST1 circuit drives (i) expression of the stem cell markers SOX2, BMI1, ALDH1, and CD44, (ii) self-renewal capacity, and (iii) tumorigenicity. In concert with these results, we show that MUC1-C and TWIST1 also drive EMT and stemness in association with acquired paclitaxel (PTX) resistance. Of potential therapeutic importance, targeting MUC1-C and thereby TWIST1 reverses the PTX refractory phenotype as evidenced by synergistic activity with PTX against drug-resistant cells. These findings uncover a master role for MUC1-C in driving the induction of TWIST1, EMT, stemness, and drug resistance, and support MUC1-C as a highly attractive target for inhibiting TNBC plasticity and progression.
Collapse
Affiliation(s)
- Tsuyoshi Hata
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Yamamoto
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Caining Jin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yan Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ling Kui
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yota Yasumizu
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Deli Hong
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Miyo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masayuki Hiraki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Takahiro Maeda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yozo Suzuki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hidekazu Takahashi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mehmet Samur
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
32
|
Baulida J, Díaz VM, Herreros AGD. Snail1: A Transcriptional Factor Controlled at Multiple Levels. J Clin Med 2019; 8:jcm8060757. [PMID: 31141910 PMCID: PMC6616578 DOI: 10.3390/jcm8060757] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/27/2022] Open
Abstract
Snail1 transcriptional factor plays a key role in the control of epithelial to mesenchymal transition and fibroblast activation. As a consequence, Snail1 expression and function is regulated at multiple levels from gene transcription to protein modifications, affecting its interaction with specific cofactors. In this review, we describe the different elements that control Snail1 expression and its activity both as transcriptional repressor or activator.
Collapse
Affiliation(s)
- Josep Baulida
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unidad Asociada al CSIC, 08003 Barcelona, Spain.
| | - Víctor M Díaz
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unidad Asociada al CSIC, 08003 Barcelona, Spain.
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003 Barcelona, Spain.
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unidad Asociada al CSIC, 08003 Barcelona, Spain.
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08003 Barcelona, Spain.
| |
Collapse
|
33
|
EMT is associated with an epigenetic signature of ECM remodeling genes. Cell Death Dis 2019; 10:205. [PMID: 30814494 PMCID: PMC6393505 DOI: 10.1038/s41419-019-1397-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Type III epithelial–mesenchymal transition (EMT) has been previously associated with increased cell migration, invasion, metastasis, and therefore cancer aggressiveness. This reversible process is associated with an important gene expression reprogramming mainly due to epigenetic plasticity. Nevertheless, most of the studies describing the central role of epigenetic modifications during EMT were performed in a single-cell model and using only one mode of EMT induction. In our study, we studied the overall modulations of gene expression and epigenetic modifications in four different EMT-induced cell models issued from different tissues and using different inducers of EMT. Pangenomic analysis (transcriptome and ChIP–sequencing) validated our hypothesis that gene expression reprogramming during EMT is largely regulated by epigenetic modifications of a wide range of genes. Indeed, our results confirmed that each EMT model is unique and can be associated with a specific transcriptome profile and epigenetic program. However, we could select some genes or pathways that are similarly regulated in the different models and that could therefore be used as a common signature of all EMT models and become new biomarkers of the EMT phenotype. As an example, we can cite the regulation of gene-coding proteins involved in the degradation of the extracellular matrix (ECM), which are highly induced in all EMT models. Based on our investigations and results, we identified ADAM19 as a new biomarker of in vitro and in vivo EMT and we validated this biological new marker in a cohort of non-small lung carcinomas.
Collapse
|
34
|
Deng M, Qin Y, Chen X, Wang Q, Wang J. MiR-206 inhibits proliferation, migration, and invasion of gastric cancer cells by targeting the MUC1 gene. Onco Targets Ther 2019; 12:849-859. [PMID: 30774372 PMCID: PMC6355168 DOI: 10.2147/ott.s180021] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background MicroRNAs (miRNAs) can regulate the post-transcriptional level of gene expression. It has been documented that downregulation of miR-206 is significant in human gastric cancer (GC), whereas its role in GC cell biological behaviors, including proliferation, migration, and invasion, has not been thoroughly investigated. MiR-206 levels have a negative association with lymph node metastasis and tumor invasion, and patients with higher miR-206 expression have better prognoses. Functional studies demonstrated that miR-206 overexpression significantly suppresses GC cell proliferation, migration, and invasion, and induces apoptosis in vitro. Materials and methods MiR-206 and MUC1 were determined by RNA extraction, quantitative real-time polymerase chain reaction, and luciferase reporter gene assays. The viability of GC cells was tested using the Cell Counting Kit 8 assay. Transwell invasion and migration assays detected GC cancer cell proliferation, invasion, and migration. Flow cytometry was applied to analyze apoptotic cells. FACS analysis was applied to detect the mitochondrial membrane potential of cells. Western blotting assay determined protein levels. Results The luciferase reporter gene assay demonstrated that miR-206 might directly bind to the 3'UTR of the MUC1 gene and suppress MUC1 expression. Furthermore, MUCI expression was upregulated and inversely associated with miR-206 levels in GC tissues. More importantly, the miR-206-mediated suppression of proliferation, migration, and invasion, and the induction of apoptosis, were abrogated by MUC1 overexpression. Conclusion Our data demonstrated that miR-206 may exert antitumor activities through inhibiting the expression of MUC1, which may serve as an effective and potential target for GC treatment.
Collapse
Affiliation(s)
- Min Deng
- Department of Gastroenterology, The First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui 233004, People's Republic of China
| | - Yiyu Qin
- Clinical Medical College, Research Centre of Biomedical Technology, Yancheng Institute of Health Sciences, Yancheng, Jiangsu 224005, People's Republic of China
| | - Xiaodong Chen
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, People's Republic of China,
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui 233004, People's Republic of China
| | - Jianchao Wang
- Department of Gastroenterology, The First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui 233004, People's Republic of China
| |
Collapse
|
35
|
Guo M, You C, Dou J. Role of transmembrane glycoprotein mucin 1 (MUC1) in various types of colorectal cancer and therapies: Current research status and updates. Biomed Pharmacother 2018; 107:1318-1325. [PMID: 30257347 DOI: 10.1016/j.biopha.2018.08.109] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal carcinoma (CRC) is the third most common malignant tumor in the world. In recent years, the morbidity and mortality of CRC have increased in the world due to increasingly ageing population, modern dietary habits, environmental change, genetic disorders and chronic intestinal inflammation. Despite recent advances in earlier detection and improvements in chemotherapy, the 5-year survival rate of patients with metastatic CRC remains low. Therefore, novel effective treatment strategies for primary or metastatic CRC have emerged to enhance cure rate as well as elongation of patient's survival. Immunotherapy has been proposed for a potentially effective therapeutic approach to the treatment of CRC. Tumor vaccination in preclinical and clinical studies has supported the antitumor activity induced by immunization with CRC cell vaccines. Epithelial cell molecule Mucin 1 (MUC1), a transmembrane glycoprotein aberrantly overexpressed in various cancers including CRC, has been used as a candidate target antigen in the peptide, dendritic cell, and whole tumor vaccines. Several clinical trials in progress reveal the immunogenicity and suitability of MUC1 that acted as immunotherapeutic vaccines for CRC/colorectal cancer stem cells (CCSC). The present review summarizes the potential roles of MUC1 on CRC/CCSC vaccines according to the latest data. Moreover, this review also discusses the novel strategies for targeting CCSC via inducing an immune response against MUC1 to achieve the best prevention and treatment effects in animal models and clinical trails.
Collapse
Affiliation(s)
- Mei Guo
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Chengzhong You
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
36
|
Dong Q, Zhang H, Han Y, Djamila A, Cheng H, Tang Z, Zhou J, Ding Y. Tumor environment differentiated “nanodepot” programmed for site-specific drug shuttling and combinative therapy on metastatic cancer. J Control Release 2018; 283:59-75. [DOI: 10.1016/j.jconrel.2018.05.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 12/31/2022]
|
37
|
Ding D, Li C, Zhao T, Li D, Yang L, Zhang B. LncRNA H19/miR-29b-3p/PGRN Axis Promoted Epithelial-Mesenchymal Transition of Colorectal Cancer Cells by Acting on Wnt Signaling. Mol Cells 2018; 41:423-435. [PMID: 29754471 PMCID: PMC5974619 DOI: 10.14348/molcells.2018.2258] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/30/2017] [Accepted: 01/14/2018] [Indexed: 02/07/2023] Open
Abstract
This investigation was aimed at working out the combined role of lncRNA H19, miR-29b and Wnt signaling in the development of colorectal cancer (CRC). In the aggregate, 185 CRC tissues and corresponding para-carcinoma tissues were gathered. The human CRC cell lines (i.e. HT29, HCT116, SW480 and SW620) and normal colorectal mucosa cell line (NCM460) were also purchased. Si-H19, si-NC, miR-29b-3p mimics, miR-29b-3p inhibitor, si-PGRN and negative control (NC) were, respectively, transfected into the CRC cells. Lucif-erase reporter plasmids were prepared to evaluate the transduction activity of Wnt/β-catenin signaling pathway, and dual-luciferase reporter gene assay was arranged to confirm the targeted relationship between H19 and miR-29b-3p, as well as between miR-29b-3p and PGRN. Finally, the proliferative and invasive capacities of CRC cells were appraised through transwell, MTT and scratch assays. As a result, over-expressed H19 and down-expressed miR-29b-3p displayed close associations with the CRC patients' poor prognosis (P < 0.05). Besides, transfection with si-H19, miR-29b-3p mimic or si-PGRN were correlated with elevated E-cadherin expression, decreased snail and vimentin expressions, as well as less-motivated cell proliferation and cell metastasis (P < 0.05). Moreover, H19 was verified to directly target miR-29b-3p based on the luciferase reporter gene assay (P < 0.05), and miR-29b-3p also bound to PGRN in a direct manner (P < 0.05). Finally, addition of LiCl (Wnt/β-catenin pathway activator) or XAV93920 (Wnt/β-catenin pathway inhibitor) would cause remarkably altered E-cadherin, c-Myc, vimentin and snail expressions, as well as significantly changed transcriptional activity of β-catenin/Tcf reporter plasmid (P < 0.05). In conclusion, the lncRNA H19/miR-29b-3p/PGRN/Wnt axis counted a great deal for seeking appropriate diagnostic biomarkers and treatment targets for CRC.
Collapse
Affiliation(s)
- Dayong Ding
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| | - Tiancheng Zhao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| | - Dandan Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| | - Lei Yang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| | - Bin Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033,
P.R. China
| |
Collapse
|
38
|
Katoh M. Multi‑layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β‑catenin signaling activation (Review). Int J Mol Med 2018; 42:713-725. [PMID: 29786110 PMCID: PMC6034925 DOI: 10.3892/ijmm.2018.3689] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
β-catenin/CTNNB1 is an intracellular scaffold protein that interacts with adhesion molecules (E-cadherin/CDH1, N-cadherin/CDH2, VE-cadherin/CDH5 and α-catenins), transmembrane-type mucins (MUC1/CD227 and MUC16/CA125), signaling regulators (APC, AXIN1, AXIN2 and NHERF1/EBP50) and epigenetic or transcriptional regulators (BCL9, BCL9L, CREBBP/CBP, EP300/p300, FOXM1, MED12, SMARCA4/BRG1 and TCF/LEF). Gain-of-function CTTNB1 mutations are detected in bladder cancer, colorectal cancer, gastric cancer, liver cancer, lung cancer, pancreatic cancer, prostate cancer and uterine cancer, whereas loss-of-function CTNNB1 mutations are also detected in human cancer. ABCB1, ALDH1A1, ASCL2, ATF3, AXIN2, BAMBI, CCND1, CD44, CLDN1, CTLA4, DKK1, EDN1, EOMES, FGF18, FGF20, FZD7, IL10, JAG1, LEF1, LGR5, MITF, MSX1, MYC, NEUROD1, NKD1, NODAL, NOTCH2, NOTUM, NRCAM, OPN, PAX3, PPARD, PTGS2, RNF43, SNAI1, SP5, TCF7, TERT, TNFRSF19, VEGFA and ZNRF3 are representative β-catenin target genes. β-catenin signaling is involved in myofibroblast activation and subsequent pulmonary fibrosis, in addition to other types of fibrosis. β-catenin and NF-κB signaling activation are involved in field cancerization in the stomach associated with Helicobacter pylori (H. pylori) infection and in the liver associated with hepatitis C virus (HCV) infection and other etiologies. β-catenin-targeted therapeutics are functionally classified into β-catenin inhibitors targeting upstream regulators (AZ1366, ETC-159, G007-LK, GNF6231, ipafricept, NVP-TNKS656, rosmantuzumab, vantictumab, WNT-C59, WNT974 and XAV939), β-catenin inhibitors targeting protein-protein interactions (CGP049090, CWP232228, E7386, ICG-001, LF3 and PRI-724), β-catenin inhibitors targeting epigenetic regulators (PKF118-310), β-catenin inhibitors targeting mediator complexes (CCT251545 and cortistatin A) and β-catenin inhibitors targeting transmembrane-type transcriptional outputs, including CD44v6, FZD7 and LGR5. Eradicating H. pylori and HCV is the optimal approach for the first-line prevention of gastric cancer and hepatocellular carcinoma (HCC), respectively. However, β-catenin inhibitors may be applicable for the prevention of organ fibrosis, second-line HCC prevention and treating β-catenin-driven cancer. The multi-layered prevention and treatment strategy of β-catenin-related human diseases is necessary for the practice of personalized medicine and implementation of precision medicine.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Chuo Ward, Tokyo 104‑0045, Japan
| |
Collapse
|
39
|
Li YL, Jin YF, Liu XX, Li HJ. A comprehensive analysis of Wnt/β-catenin signaling pathway-related genes and crosstalk pathways in the treatment of As 2O 3 in renal cancer. Ren Fail 2018; 40:331-339. [PMID: 29633893 PMCID: PMC6014489 DOI: 10.1080/0886022x.2018.1456461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We aimed to investigate the effect of As2O3 treatment on Wnt/β-catenin signaling pathway-related genes and pathways in renal cancer. Illumina-based RNA-seq of 786-O cells with or without As2O3 treatment was performed, and differentially expressed genes (DEGs) were identified using Cuffdiff software. TargetMine was utilized to perform Gene Ontology (GO) pathway and Disease Ontology enrichment analyses. Furthermore, TRANSFAC database and LPIA method were applied to select differentially expressed transcription factors (TFs) and pathways related to Wnt/β-catenin signaling pathway, respectively. Additionally, transcriptional regulatory and pathway crosstalk networks were constructed. In total, 1684 DEGs and 69 TFs were screened out. The 821 up-regulated DEGs were mainly enriched in 67 pathways, 70 GO terms, and 46 disease pathways, while only 1 pathway and 5 GO terms were enriched for 863 down-regulated DEGs. A total of 18 DEGs (4 up-regulated and 14 down-regulated genes) were involved in the Wnt/β-catenin signaling pathway. Among the 18 DEGs, 4 ones were TFs. Furthermore, 211 pathways were predicted to be linked to the Wnt/β-catenin signaling pathway. In conclusion, As2O3 may have a significant effect on the Wnt/β-catenin signaling pathway for renal cancer treatment. The potential key DEGs are expected to be used as therapeutic targets.
Collapse
Affiliation(s)
- Yan-Lei Li
- a Medical Examination Center , China-Japan Union Hospital of Jilin University , Changchun , China
| | - Yu-Fen Jin
- b Clinical Laboratory , The Second Hospital of Jilin University , Changchun , China
| | - Xiu-Xia Liu
- b Clinical Laboratory , The Second Hospital of Jilin University , Changchun , China
| | - Hong-Jun Li
- a Medical Examination Center , China-Japan Union Hospital of Jilin University , Changchun , China
| |
Collapse
|
40
|
Yang X, Zhu S, Li L, Zhang L, Xian S, Wang Y, Cheng Y. Identification of differentially expressed genes and signaling pathways in ovarian cancer by integrated bioinformatics analysis. Onco Targets Ther 2018; 11:1457-1474. [PMID: 29588600 PMCID: PMC5858852 DOI: 10.2147/ott.s152238] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background The mortality rate associated with ovarian cancer ranks the highest among gynecological malignancies. However, the cause and underlying molecular events of ovarian cancer are not clear. Here, we applied integrated bioinformatics to identify key pathogenic genes involved in ovarian cancer and reveal potential molecular mechanisms. Results The expression profiles of GDS3592, GSE54388, and GSE66957 were downloaded from the Gene Expression Omnibus (GEO) database, which contained 115 samples, including 85 cases of ovarian cancer samples and 30 cases of normal ovarian samples. The three microarray datasets were integrated to obtain differentially expressed genes (DEGs) and were deeply analyzed by bioinformatics methods. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments of DEGs were performed by DAVID and KOBAS online analyses, respectively. The protein–protein interaction (PPI) networks of the DEGs were constructed from the STRING database. A total of 190 DEGs were identified in the three GEO datasets, of which 99 genes were upregulated and 91 genes were downregulated. GO analysis showed that the biological functions of DEGs focused primarily on regulating cell proliferation, adhesion, and differentiation and intracellular signal cascades. The main cellular components include cell membranes, exosomes, the cytoskeleton, and the extracellular matrix. The molecular functions include growth factor activity, protein kinase regulation, DNA binding, and oxygen transport activity. KEGG pathway analysis showed that these DEGs were mainly involved in the Wnt signaling pathway, amino acid metabolism, and the tumor signaling pathway. The 17 most closely related genes among DEGs were identified from the PPI network. Conclusion This study indicates that screening for DEGs and pathways in ovarian cancer using integrated bioinformatics analyses could help us understand the molecular mechanism underlying the development of ovarian cancer, be of clinical significance for the early diagnosis and prevention of ovarian cancer, and provide effective targets for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Obstetrics and Gynecology
| | - Shaoming Zhu
- Department of Urology, Renmin Hospital of Wuhan University
| | - Li Li
- sDepartment of Pharmacology, Wuhan University Health Science Center, Wuhan, Hubei, People's Republic of China
| | - Li Zhang
- Department of Obstetrics and Gynecology
| | - Shu Xian
- Department of Obstetrics and Gynecology
| | | | | |
Collapse
|
41
|
Xu Z, Liu Y, Yang Y, Wang J, Zhang G, Liu Z, Fu H, Wang Z, Liu H, Xu J. High expression of Mucin13 associates with grimmer postoperative prognosis of patients with non-metastatic clear-cell renal cell carcinoma. Oncotarget 2018; 8:7548-7558. [PMID: 27911274 PMCID: PMC5352342 DOI: 10.18632/oncotarget.13692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/22/2016] [Indexed: 01/11/2023] Open
Abstract
Background Mucin13 (MUC13) is a transmembrane glycoprotein that is aberrantly expressed in ovarian and gastro-intestinal tumors, but its role in renal cell carcinoma remains elusive. The purpose of this study is to evaluate the prognostic value of MUC13 expression in patients with non-metastatic clear cell renal cell carcinoma (ccRCC) after surgical resection. Results MUC13 high expression was associated with high Fuhrman grade (p < 0.001), high SSIGN score (p = 0.011), early recurrence (p < 0.001) and poor survival (p < 0.001). Multivariate Cox regression analysis identified MUC13 expression as an independent prognostic factor for RFS and OS of ccRCC patients. A nomogram integrating MUC13 expression and other independent prognosticators was established to predict RFS and OS of ccRCC patients. Optimal agreement was shown between the predictions and observations in calibration curves. Matrials and methods This study enrolled 410 postoperative non-metastatic ccRCC patients at a single institution. Clinicopathologic variables, recurrence-free survival (RFS), and overall survival (OS) were recorded. MUC13 expression was detected by immunohistochemical staining in tumor specimens. Association of MUC13 expression with clinicopathological factors was explored. Kaplan-Meier analysis was performed to compare survival curves. Univariate and multivariate Cox regression models were used to analyze the impact of prognostic factors on RFS and OS. A prognostic nomogram was constructed based on the independent prognostic factors identified by multivariate analysis. Conclusions MUC13 high expression is a novel independent adverse prognostic factor of clinical outcome in non-metastatic ccRCC patients after surgery.
Collapse
Affiliation(s)
- Zhiying Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yidong Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanfeng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jieti Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Guodong Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zheng Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Hangcheng Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zewei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
42
|
C-terminus of MUC16 activates Wnt signaling pathway through its interaction with β-catenin to promote tumorigenesis and metastasis. Oncotarget 2018; 7:36800-36813. [PMID: 27167110 PMCID: PMC5095040 DOI: 10.18632/oncotarget.9191] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/18/2016] [Indexed: 12/14/2022] Open
Abstract
MUC16/CA125 has been identified as a prominent cancer biomarker, especially for epithelial ovarian cancers, in clinical test for over three decades. Due to its huge mass, limited knowledge of MUC16 was acquired previously. By utilizing a well characterized self-made MUC16 monoclonal antibody, we identified the endogenous interaction between a C-terminal fragment of MUC16 (MUC16C) and β-catenin for the first time, and further elucidated that trans-activation domain of β-catenin is required for this interaction. Such interaction could activate the Wnt/β-catenin signaling pathway by facilitating cytosol-nucleus transportation of β-catenin, consequently induce cell proliferation and the migration, eventually lead to tumorigenesis and metastasis in nude mice. Consistently, knockdown of MUC16 significantly weakened the capabilities of cells for proliferation and migration. Based on our discovery, we suggest that MUC16 appears as an attractive target for the development of effective anticancer drugs.
Collapse
|
43
|
Depletion of MUC5B mucin in gastrointestinal cancer cells alters their tumorigenic properties: implication of the Wnt/β-catenin pathway. Biochem J 2017; 474:3733-3746. [PMID: 28972071 DOI: 10.1042/bcj20170348] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022]
Abstract
Secreted mucins are large O-glycosylated proteins that participate in the protection/defence of underlying mucosae in normal adults. Alteration of their expression is a hallmark of numerous epithelial cancers and has often been correlated to bad prognosis of the tumour. The secreted mucin MUC5B is overexpressed in certain subtypes of gastric and intestinal cancers, but the consequences of this altered expression on the cancer cell behaviour are not known. To investigate the role of MUC5B in carcinogenesis, its expression was knocked-down in the human gastric cancer cell line KATO-III and in the colonic cancer cell line LS174T by using transient and stable approaches. Consequences of MUC5B knocking-down on cancer cells were studied with respect to in vitro proliferation, migration and invasion, and in vivo on tumour growth using a mouse subcutaneous xenograft model. Western blotting, luciferase assay and qRT-PCR were used to identify proteins and signalling pathways involved. In vitro MUC5B down-regulation leads to a decrease in proliferation, migration and invasion properties in both cell lines. Molecular mechanisms involved the alteration of β-catenin expression, localization and activity and decreased expression of several of its target genes. In vivo xenografts of MUC5B-deficient cells induced a decrease in tumour growth when compared with MUC5B-expressing Mock cells. Altogether, the present study shows that down-regulation of MUC5B profoundly alters proliferation, migration and invasion of human gastrointestinal cancer cells and that these alterations may be, in part, mediated by the Wnt/β-catenin pathway emphasizing the potential of MUC5B as an actor of gastrointestinal carcinogenesis.
Collapse
|
44
|
Growth-induced stress enhances epithelial-mesenchymal transition induced by IL-6 in clear cell renal cell carcinoma via the Akt/GSK-3β/β-catenin signaling pathway. Oncogenesis 2017; 6:e375. [PMID: 28846080 PMCID: PMC5608922 DOI: 10.1038/oncsis.2017.74] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/24/2017] [Accepted: 07/07/2017] [Indexed: 12/18/2022] Open
Abstract
Stromal cell populations in the tumor microenvironment (TME) play a critical role in the oncogenesis and metastasis of renal cell carcinoma. In this study, we found that there are α-smooth muscle actin positive (α-SMA (+)) cells in the stroma of clear cell renal cell carcinoma (ccRCC) tissues, and their numbers are significantly associated with poor survival in ccRCC patients. Interleukin 6 (IL-6) is a critical diver that induces α-SMA (+) cells in ccRCC tissues via promotion of epithelial to mesenchymal transition (EMT) and stimulates migration and invasion in ccRCC. Peritumoral CD4+ T cells are the main source of IL-6 in ccRCC tissues. In addition to biochemical factors, mechanical compression within tumors affects tumor cell behavior. Tumors grown in a confined space exhibit intratumoral compressive stress and, with sufficient pressure, stress-stimulated migration of cancer cells. Moreover, a combination of IL-6 secreted by CD4+ T cells and growth-induced solid stress further contributes to the regulation of cancer cell morphogenesis, EMT and acquisition of a stemness phenotype. The effects in the combination group were driven by the Akt/GSK-3β/β-catenin signaling pathway, and deregulation of β-catenin expression was predictive of poor outcome in ccRCC patients. Notably, the expression of a cancer stem cell marker, CD44, was correlated with T stage, high Fuhrman grade and metastasis in ccRCC. These data provide evidence for new stress-reducing and IL-6 targeting strategies in cancer therapy.
Collapse
|
45
|
Gaudelot K, Gibier JB, Pottier N, Hémon B, Van Seuningen I, Glowacki F, Leroy X, Cauffiez C, Gnemmi V, Aubert S, Perrais M. Targeting miR-21 decreases expression of multi-drug resistant genes and promotes chemosensitivity of renal carcinoma. Tumour Biol 2017; 39:1010428317707372. [PMID: 28714373 DOI: 10.1177/1010428317707372] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Renal cell carcinoma, the most common neoplasm of adult kidney, accounts for about 3% of adult malignancies and is usually highly resistant to conventional therapy. MicroRNAs are a class of small non-coding RNAs, which have been previously shown to promote malignant initiation and progression. In this study, we focused our attention on miR-21, a well described oncomiR commonly upregulated in cancer. Using a cohort of 99 primary renal cell carcinoma samples, we showed that miR-21 expression in cancer tissues was higher than in adjacent non-tumor tissues whereas no significant difference was observed with stages, grades, and metastatic outcome. In vitro, miR-21 was also overexpressed in renal carcinoma cell lines compared to HK-2 human proximal tubule epithelial cell line. Moreover, using Boyden chambers and western blot techniques, we also showed that miR-21 overexpression increased migratory, invasive, proliferative, and anti-apoptotic signaling pathways whereas opposite results were observed using an anti-miR-21-based silencing strategy. Finally, we assessed the role of miR-21 in mediating renal cell carcinoma chemoresistance and further showed that miR-21 silencing significantly (1) increased chemosensitivity of paclitaxel, 5-fluorouracil, oxaliplatin, and dovitinib; (2) decreased expression of multi-drug resistance genes; and (4) increased SLC22A1/OCT1, SLC22A2/OCT2, and SLC31A1/CTR1 platinum influx transporter expression. In conclusion, our results showed that miR-21 is a key actor of renal cancer progression and plays an important role in the resistance to chemotherapeutic drugs. In renal cell carcinoma, targeting miR-21 is a potential new therapeutic strategy to improve chemotherapy efficacy and consequently patient outcome.
Collapse
Affiliation(s)
- Kelly Gaudelot
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France
| | - Jean-Baptiste Gibier
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France.,2 CHU Lille, Pathology Institute, Centre de Biologie Pathologie, Rue Oscar Lambret, Lille, France
| | - Nicolas Pottier
- 3 EA4483, Université de Lille, Faculté de Médecine, Pôle Recherche, Place de Verdun, Lille, France.,4 CHU Lille, Department of Biochemistry and Molecular Biology, Centre de Biologie Pathologie, Rue Oscar Lambret, Lille, France
| | - Brigitte Hémon
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France
| | - Isabelle Van Seuningen
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France
| | - François Glowacki
- 3 EA4483, Université de Lille, Faculté de Médecine, Pôle Recherche, Place de Verdun, Lille, France.,5 CHU Lille, Department of Nephrology, Hôpital Huriez, Rue Michel Polonovski, Lille, France
| | - Xavier Leroy
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France.,2 CHU Lille, Pathology Institute, Centre de Biologie Pathologie, Rue Oscar Lambret, Lille, France
| | - Christelle Cauffiez
- 3 EA4483, Université de Lille, Faculté de Médecine, Pôle Recherche, Place de Verdun, Lille, France
| | - Viviane Gnemmi
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France.,2 CHU Lille, Pathology Institute, Centre de Biologie Pathologie, Rue Oscar Lambret, Lille, France
| | - Sébastien Aubert
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France.,2 CHU Lille, Pathology Institute, Centre de Biologie Pathologie, Rue Oscar Lambret, Lille, France
| | - Michaël Perrais
- 1 Université de Lille, Inserm, CHU Lille, UMR-S 1172, Team "Mucins, Epithelial Differentiation and Carcinogenesis," Jean-Pierre Aubert Research Center (JPARC), Lille, France
| |
Collapse
|
46
|
Gibier JB, Hémon B, Fanchon M, Gaudelot K, Pottier N, Ringot B, Van Seuningen I, Glowacki F, Cauffiez C, Blum D, Copin MC, Perrais M, Gnemmi V. Dual role of MUC1 mucin in kidney ischemia-reperfusion injury: Nephroprotector in early phase, but pro-fibrotic in late phase. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1336-1349. [DOI: 10.1016/j.bbadis.2017.03.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/14/2017] [Accepted: 03/29/2017] [Indexed: 11/15/2022]
|
47
|
Khaniabadi PM, Majid AMS, Asif M, Khaniabadi BM, Shahbazi-Gahrouei D, Jaafar MS. Breast cancer cell targeted MR molecular imaging probe: Anti-MUC1 antibody-based magnetic nanoparticles. ACTA ACUST UNITED AC 2017. [DOI: 10.1088/1742-6596/851/1/012014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
48
|
Bouillez A, Rajabi H, Jin C, Samur M, Tagde A, Alam M, Hiraki M, Maeda T, Hu X, Adeegbe D, Kharbanda S, Wong KK, Kufe D. MUC1-C integrates PD-L1 induction with repression of immune effectors in non-small-cell lung cancer. Oncogene 2017; 36:4037-4046. [PMID: 28288138 PMCID: PMC5509481 DOI: 10.1038/onc.2017.47] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/15/2016] [Accepted: 02/01/2017] [Indexed: 12/20/2022]
Abstract
Immunotherapeutic approaches, particularly PD-1/PD-L1 blockade, have improved the treatment of non-small cell lung cancer (NSCLC), supporting the premise that evasion of immune destruction is of importance for NSCLC progression. However, the signals responsible for upregulation of PD-L1 in NSCLC cells and whether they are integrated with the regulation of other immune-related genes are not known. Mucin 1 (MUC1) is aberrantly overexpressed in NSCLC, activates the NF-κB p65→ZEB1 pathway and confers a poor prognosis. The present studies demonstrate that MUC1-C activates PD-L1 expression in NSCLC cells. We show that MUC1-C increases NF-κB p65 occupancy on the CD274/PD-L1 promoter and thereby drives CD274 transcription. Moreover, we demonstrate that MUC1-C-induced activation of NF-κB→ZEB1 signaling represses the TLR9, IFNG, MCP-1 and GM-CSF genes, and that this signature is associated with decreases in overall survival. In concert with these results, targeting MUC1-C in NSCLC tumors suppresses PD-L1 and induces these effectors of innate and adaptive immunity. These findings support a previously unrecognized central role for MUC1-C in integrating PD-L1 activation with suppression of immune effectors and poor clinical outcome.
Collapse
Affiliation(s)
- A Bouillez
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - H Rajabi
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - C Jin
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Samur
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - A Tagde
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Alam
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Hiraki
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - T Maeda
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - X Hu
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Adeegbe
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - S Kharbanda
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - K-K Wong
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Kufe
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Rajabi H, Tagde A, Alam M, Bouillez A, Pitroda S, Suzuki Y, Kufe D. DNA methylation by DNMT1 and DNMT3b methyltransferases is driven by the MUC1-C oncoprotein in human carcinoma cells. Oncogene 2016; 35:6439-6445. [PMID: 27212035 PMCID: PMC5121097 DOI: 10.1038/onc.2016.180] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 12/23/2022]
Abstract
Aberrant expression of the DNA methyltransferases (DNMTs) and disruption of DNA methylation patterns are associated with carcinogenesis and cancer cell survival. The oncogenic MUC1-C protein is aberrantly overexpressed in diverse carcinomas; however, there is no known link between MUC1-C and DNA methylation. Our results demonstrate that MUC1-C induces the expression of DNMT1 and DNMT3b, but not DNMT3a, in breast and other carcinoma cell types. We show that MUC1-C occupies the DNMT1 and DNMT3b promoters in complexes with NF-κB p65 and drives DNMT1 and DNMT3b transcription. In this way, MUC1-C controls global DNA methylation as determined by analysis of LINE-1 repeat elements. The results further demonstrate that targeting MUC1-C downregulates DNA methylation of the CDH1 tumor suppressor gene in association with induction of E-cadherin expression. These findings provide compelling evidence that MUC1-C is of functional importance to induction of DNMT1 and DNMT3b and, in turn, changes in DNA methylation patterns in cancer cells.
Collapse
Affiliation(s)
- H Rajabi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - A Tagde
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M Alam
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - A Bouillez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - S Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Y Suzuki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D Kufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Epithelial to Mesenchymal Transition in Renal Cell Carcinoma: Implications for Cancer Therapy. Mol Diagn Ther 2016; 20:111-7. [PMID: 26940073 DOI: 10.1007/s40291-016-0192-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a developmentally vital reversible process by which fully differentiated cells lose their epithelial features and acquire a migratory mesenchymal phenotype. EMT contributes to the metastatic potential of tumors. The expression profile and other biological properties of EMT suggest potential targets for cancer therapy, including in renal-cell carcinoma (RCC). The preclinical and clinical results have substantiated the promises that dysregulated elements leading to EMT can be a potential target in RCC patients. In this study, we illustrated the pathogenic and prognostic role of EMT in RCC. In addition, we reconstructed, by literature analysis, the different pathways implicated in the EMT process, thus supporting the rational for future EMT-directed therapeutic approaches for RCC patients.
Collapse
|