1
|
Li M, Wang Y, Lin X, Yang H, Zhang X, Bai Y, Li X, Zhang L, Cheng F, Cao C, Zhou Q. Evaluation of antitumor potential of an anti-glypican-1 monoclonal antibody in preclinical lung cancer models reveals a distinct mechanism of action. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:600-626. [PMID: 38966167 PMCID: PMC11220310 DOI: 10.37349/etat.2024.00238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/26/2024] [Indexed: 07/06/2024] Open
Abstract
Aim The main objective of this study was to investigate the antitumor effect of a mouse anti-human glypican-1 (GPC1) monoclonal antibody (mAb) on non-small cell lung carcinoma (NSCLC) and associated molecular mechanisms. Methods The anti-proliferative and anti-migratory activities of anti-GPC1 mAb were examined in A549 and H460 NSCLC cells and LL97A lung fibroblasts. The inhibitory effect of anti-GPC1 mAb on tumor growth was evaluated in an orthotopic lung tumor model. Results The in vitro study showed that anti-GPC1 mAb profoundly inhibited the anchorage-independent growth of A549 and H460 NSCLC cells and exhibited relatively high cytotoxic activities towards LL97A lung fibroblasts, A549/LL97A and H460/LL97A coculture spheroids. Moreover, anti-GPC1 mAb significantly decreased the expression of phospho-Src (p-Src; Tyr416), p-Akt (Ser473) and β-catenin in the co-cultured LL97A lung fibroblasts, and the expression of phospho-mitogen-activated protein kinase kinase (p-MEK; Ser217/221) and phospho-90 kDa ribosomal s6 kinase (p-p90RSK; Ser380) in co-cultured A549 cells. When anti-GPC1 mAb was administered to tumor-bearing mice, the inhibitory effect of anti-GPC1 mAb on the orthotopic lung tumor growth was not statistically significant. Nonetheless, results of Western blot analysis showed significant decrease in the phosphorylation of fibroblast growth factor receptor 1 (FGFR1) at Tyr766, Src at Tyr416, extracellular signal-regulated kinase (ERK) at Thr202/Tyr204, 90 kDa ribosomal S6 kinase (RSK) at Ser380, glycogen synthase kinases 3α (GSK3α) at Ser21 and GSK3β at Ser9 in tumor tissues. These data implicate that anti-GPC1 mAb treatment impairs the interaction between tumor cells and tumor associated fibroblasts by attenuating the paracrine FGFR signal transduction. Conclusions The relatively potent cytotoxicity of anti-GPC1 mAb in lung fibroblasts and its potential inhibitory effect on the paracrine FGFR signal transduction warrant further studies on the combined use of this mAb with targeted therapeutics to improve therapeutic outcomes in lung cancer.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yanhong Wang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaoyang Lin
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Haiqiang Yang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Xiaolin Zhang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Yun Bai
- MegaNano Biotech Inc., Tampa, FL 33612, USA
| | - Xiankun Li
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Lulu Zhang
- Zhengzhou Molecular Diagnosis Engineering Technology Research Center, Zhengzhou 450001, Henan Province, China
| | - Feng Cheng
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Qingyu Zhou
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Liguori GL. Challenges and Promise for Glioblastoma Treatment through Extracellular Vesicle Inquiry. Cells 2024; 13:336. [PMID: 38391949 PMCID: PMC10886570 DOI: 10.3390/cells13040336] [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: 12/15/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Glioblastoma (GB) is a rare but extremely aggressive brain tumor that significantly impacts patient outcomes, affecting both duration and quality of life. The protocol established by Stupp and colleagues in 2005, based on radiotherapy and chemotherapy with Temozolomide, following maximum safe surgical resection remains the gold standard for GB treatment; however, it is evident nowadays that the extreme intratumoral and intertumoral heterogeneity, as well as the invasiveness and tendency to recur, of GB are not compatible with a routine and unfortunately ineffective treatment. This review article summarizes the main challenges in the search for new valuable therapies for GB and focuses on the impact that extracellular vesicle (EV) research and exploitation may have in the field. EVs are natural particles delimited by a lipidic bilayer and filled with functional cellular content that are released and uptaken by cells as key means of cell communication. Furthermore, EVs are stable in body fluids and well tolerated by the immune system, and are able to cross physiological, interspecies, and interkingdom barriers and to target specific cells, releasing inherent or externally loaded functionally active molecules. Therefore, EVs have the potential to be ideal allies in the fight against GB and to improve the prognosis for GB patients. The present work describes the main preclinical results obtained so far on the use of EVs for GB treatment, focusing on both the EV sources and molecular cargo used in the various functional studies, primarily in vivo. Finally, a SWOT analysis is performed, highlighting the main advantages and pitfalls of developing EV-based GB therapeutic strategies. The analysis also suggests the main directions to explore to realize the possibility of exploiting EVs for the treatment of GB.
Collapse
Affiliation(s)
- Giovanna L Liguori
- Institute of Genetics and Biophysics (IGB) "Adriano Buzzati-Traverso", National Research Council (CNR) of Italy, 80131 Naples, Italy
| |
Collapse
|
3
|
Zhong J, Li L, Zhang Q, Zou J, Liu W, Xu CH. Expression and prognostic value of Cripto-1 in early non-small cell lung cancer. THE CLINICAL RESPIRATORY JOURNAL 2023; 17:1203-1208. [PMID: 37528674 PMCID: PMC10730460 DOI: 10.1111/crj.13680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/24/2023] [Accepted: 07/23/2023] [Indexed: 08/03/2023]
Abstract
OBJECTIVE We aim to explore the expression of Cripto-1 (CR-1) protein in patients with early stage non-small cell lung cancer (NSCLC). METHODS We investigated CR-1 expression status in specimens obtained from 240 patients with resected NSCLC and 30 cases of para-carcinous normal lung tissues. RESULTS Compared with normal lung tissue, the positive expression of CR-1 protein in NSCLC was significantly increased (p < 0.005). Cox multivariate regression analysis showed that the expression of CR-1 protein was an independent prognostic factor for early stage NSCLC (p = 0.002). CONCLUSION Detecting CR-1 protein can predict the prognosis and recurrence in patients with NSCLC.
Collapse
Affiliation(s)
- Jian Zhong
- Department of Thoracic SurgeryAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Li Li
- Department of Respiratory MedicineAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| | - Qian Zhang
- Department of Respiratory MedicineAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| | - Jue Zou
- Department of PathologyAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Wei Liu
- Department of Respiratory MedicineAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
| | - Chun Hua Xu
- Department of Respiratory MedicineAffiliated Nanjing Brain Hospital, Nanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| |
Collapse
|
4
|
Liguori GL, Kralj-Iglič V. Pathological and Therapeutic Significance of Tumor-Derived Extracellular Vesicles in Cancer Cell Migration and Metastasis. Cancers (Basel) 2023; 15:4425. [PMID: 37760395 PMCID: PMC10648223 DOI: 10.3390/cancers15184425] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
The infiltration of primary tumors and metastasis formation at distant sites strongly impact the prognosis and the quality of life of cancer patients. Current therapies including surgery, radiotherapy, and chemotherapy are limited in targeting the complex cell migration mechanisms responsible for cancer cell invasiveness and metastasis. A better understanding of these mechanisms and the development of new therapies are urgently needed. Extracellular vesicles (EVs) are lipid-enveloped particles involved in inter-tissue and inter-cell communication. This review article focuses on the impact of EVs released by tumor cells, specifically on cancer cell migration and metastasis. We first introduce cell migration processes and EV subtypes, and we give an overview of how tumor-derived EVs (TDEVs) may impact cancer cell migration. Then, we discuss ongoing EV-based cancer therapeutic approaches, including the inhibition of general EV-related mechanisms as well as the use of EVs for anti-cancer drug delivery, focusing on the harnessing of TDEVs. We propose a protein-EV shuttle as a route alternative to secretion or cell membrane binding, influencing downstream signaling and the final effect on target cells, with strong implications in tumorigenesis. Finally, we highlight the pitfalls and limitations of therapeutic EV exploitation that must be overcome to realize the promise of EVs for cancer therapy.
Collapse
Affiliation(s)
- Giovanna L. Liguori
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati-Traverso”, National Research Council (CNR) of Italy, 80131 Naples, Italy
| | - Veronika Kralj-Iglič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
5
|
Xu C, Zou J, Li L, Yuan Q, Wang W. Elevated serum Cripto-1 and VEGF levels in patients with non-small cell lung cancer. FASEB Bioadv 2022; 4:539-546. [PMID: 35949510 PMCID: PMC9353448 DOI: 10.1096/fba.2022-00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022] Open
Abstract
Cripto-1 (CR-1) facilitates vascular endothelial growth factor (VEGF) expression, and these markers are associated with various tumor cell proliferation, angiogenesis, and metastasis. The main aim of our study was to investigate the clinical value of CR-1 and VEGF for non-small cell lung cancer (NSCLC) patients. Serum samples were collected from 312 patients with NSCLC and 120 healthy controls. The levels of CR-1 and VEGF were measured by enzyme-linked immunosorbent assay (ELISA). The serum levels of CR-1 and VEGF in NSCLC patients were significantly higher than those of healthy controls (p < 0.05). Elevated CR-1 levels were associated with progression of NSCLC stage and higher CR-1 was detected more in patients with distant metastasis (p < 0.05). Patients exhibiting low levels of serum CR-1 had better overall survival than those with high levels (p < 0.05). The CR-1 levels of NSCLC patients with postoperative recurrent were higher than those of nonrecurrent NSCLC patients. Our study suggests that serum CR-1 and VEGF are useful biomarker for NSCLC patients.
Collapse
Affiliation(s)
- Chunhua Xu
- Department of Respiratory Medicine, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| | - Jue Zou
- Department of Respiratory Medicine, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
- Department of Pathology, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
| | - Li Li
- Department of Respiratory Medicine, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| | - Qi Yuan
- Department of Respiratory Medicine, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| | - Wei Wang
- Department of Respiratory Medicine, Affiliated Nanjing Brain HospitalNanjing Medical UniversityNanjingChina
- Clinical Center of Nanjing Respiratory Diseases and ImagingNanjingChina
| |
Collapse
|
6
|
Mantile F, Kisovec M, Adamo G, Romancino DP, Hočevar M, Božič D, Bedina Zavec A, Podobnik M, Stoppelli MP, Kisslinger A, Bongiovanni A, Kralj-Iglič V, Liguori GL. A Novel Localization in Human Large Extracellular Vesicles for the EGF-CFC Founder Member CRIPTO and Its Biological and Therapeutic Implications. Cancers (Basel) 2022; 14:cancers14153700. [PMID: 35954365 PMCID: PMC9367246 DOI: 10.3390/cancers14153700] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Tumor growth and metastasis strongly rely on cell–cell communication. One of the mechanisms by which tumor cells communicate involves the release and uptake of lipid membrane encapsulated particles full of bioactive molecules, called extracellular vesicles (EVs). EV exchange between cancer cells may induce phenotype changes in the recipient cells. Our work investigated the effect of EVs released by teratocarcinoma cells on glioblastoma (GBM) cells. EVs were isolated by differential centrifugation and analyzed through Western blot, nanoparticle tracking analysis, and electron microscopy. The effect of large EVs on GBM cells was tested through cell migration, proliferation, and drug-sensitivity assays, and resulted in a specific impairment in cell migration with no effects on proliferation and drug-sensitivity. Noticeably, we found the presence of the EGF-CFC founder member CRIPTO on both small and large EVs, in the latter case implicated in the EV-mediated negative regulation of GBM cell migration. Our data let us propose a novel route and function for CRIPTO during tumorigenesis, highlighting a complex scenario regulating its effect, and paving the way to novel strategies to control cell migration, to ultimately improve the prognosis and quality of life of GBM patients.
Collapse
Affiliation(s)
- Francesca Mantile
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati Traverso”, National Research Council (CNR) of Italy, 80131 Naples, Italy; (F.M.); (M.P.S.)
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (A.B.Z.); (M.P.)
| | - Giorgia Adamo
- Institute for Research and Biomedical Innovation (IRIB), CNR, 90146 Palermo, Italy; (G.A.); (D.P.R.); (A.B.)
| | - Daniele P. Romancino
- Institute for Research and Biomedical Innovation (IRIB), CNR, 90146 Palermo, Italy; (G.A.); (D.P.R.); (A.B.)
| | - Matej Hočevar
- Department of Physics and Chemistry of Materials, Institute of Metals and Technology, SI-1000 Ljubljana, Slovenia;
| | - Darja Božič
- Faculty of Health Sciences, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (D.B.); (V.K.-I.)
- Faculty of Electrical Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Apolonija Bedina Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (A.B.Z.); (M.P.)
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (M.K.); (A.B.Z.); (M.P.)
| | - Maria Patrizia Stoppelli
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati Traverso”, National Research Council (CNR) of Italy, 80131 Naples, Italy; (F.M.); (M.P.S.)
| | - Annamaria Kisslinger
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR) of Italy, 80131 Naples, Italy;
| | - Antonella Bongiovanni
- Institute for Research and Biomedical Innovation (IRIB), CNR, 90146 Palermo, Italy; (G.A.); (D.P.R.); (A.B.)
| | - Veronika Kralj-Iglič
- Faculty of Health Sciences, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (D.B.); (V.K.-I.)
- Faculty of Electrical Engineering, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Giovanna L. Liguori
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati Traverso”, National Research Council (CNR) of Italy, 80131 Naples, Italy; (F.M.); (M.P.S.)
- Correspondence:
| |
Collapse
|
7
|
Afify SM, Hassan G, Nawara HM, H Zahra M, Xu Y, Alam MJ, Saitoh K, Mansour H, Abu Quora HA, Sheta M, Monzur S, Du J, Oh SY, Seno A, Salomon DS, Seno M. Optimization of production and characterization of a recombinant soluble human Cripto-1 protein inhibiting self-renewal of cancer stem cells. J Cell Biochem 2022; 123:1183-1196. [PMID: 35578735 DOI: 10.1002/jcb.30272] [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: 02/26/2022] [Revised: 04/16/2022] [Accepted: 04/27/2022] [Indexed: 11/05/2022]
Abstract
Human Cripto-1 is a member of the epidermal growth factor (EGF)-Cripto-FRL-1-Cryptic (CFC) family family and performs critical roles in cancer and various pathological and developmental processes. Recently we demonstrated that a soluble form of Cripto-1 suppresses the self-renewal and enhances the differentiation of cancer stem cells (CSCs). A functional form of soluble Cripto-1 was found to be difficult to obtain because of the 12 cysteine residues in the protein which impairs the folding process. Here, we optimized the protocol for a T7 expression system, purification from inclusion bodies under denatured conditions refolding of a His-tagged Cripto-1 protein. A concentrations of 0.2-0.4 mM isopropyl β-D-1-thiogalactopyranoside (IPTG) at 37°C was found to be the optimal concentration for Cripto-1 expression while imidazole at 0.5 M was the optimum concentration to elute the Cripto-1 protein from a Ni-column in the smallest volume. Cation exchange column chromatography of the Cripto-1 protein in the presence of 8 M urea exhibited sufficient elution profile at pH 5, which was more efficient at recovery. The recovery of the protein reached to more than 26.6% after refolding with arginine. The purified Cripto-1 exhibited high affinity to the anti-ALK-4 antibody and suppressed sphere forming ability of CSCs at high dose and induced cell differentiation.
Collapse
Affiliation(s)
- Said M Afify
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Division of Biochemistry, Chemistry Department, Faculty of Science, Menoufia University, 32511, Shebin El Kom, Menofiua, Egypt
| | - Ghmkin Hassan
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Current address: Department of Genomic Oncology and Oral Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Hend M Nawara
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Maram H Zahra
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Graduate School of Natural Science and Technology, Okayama University, 7000086, okayama, okayama, Japan
| | - Yanning Xu
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Graduate School of Natural Science and Technology, Okayama University, 7000086, okayama, okayama, Japan.,Department of Pathology, Tianjin Central Hospital of Gynecology Obstetrics, Nankai University, Tianjin, China
| | - Md Jahangir Alam
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Koichi Saitoh
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Hager Mansour
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Hagar A Abu Quora
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Mona Sheta
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Department of Cancer Biology, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sadia Monzur
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - Juan Du
- Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | | | - Akimasa Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| | - David S Salomon
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Masaharu Seno
- Department of Biotechnology and Drug Discovery, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan.,Graduate School of Natural Science and Technology, Okayama University, 7000086, okayama, okayama, Japan.,Department of Cancer Stem Cell Engineering, Faculty of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 700-8530, Okayama, Japan
| |
Collapse
|
8
|
Liu Y, Li YQ, Huang SH, Li YL, Xia JW, Jia JS, Wei F, Wang JH, Dai GQ, Wang YC, Li XY, Han LX, Zhang XL, Xiang XD, Zhao WT, Xiao D, Lin XL. Liver-specific over-expression of Cripto-1 in transgenic mice promotes hepatocyte proliferation and deregulated expression of hepatocarcinogenesis-related genes and signaling pathways. Aging (Albany NY) 2021; 13:21155-21190. [PMID: 34517344 PMCID: PMC8457585 DOI: 10.18632/aging.203402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/13/2021] [Indexed: 11/25/2022]
Abstract
In this study, we investigated the role of embryonic gene Cripto-1 (CR-1) in hepatocellular carcinoma (HCC) using hepatocyte-specific CR-1-overexpressing transgenic mice. The expression of truncated 1.7-kb CR-1 transcript (SF-CR-1) was significantly higher than the full-length 2.0-kb CR-1 transcript (FL-CR-1) in a majority of HCC tissues and cell lines. Moreover, CR-1 mRNA and protein levels were significantly higher in HCC tissues than adjacent normal liver tissues. Hepatocyte-specific over-expression of CR-1 in transgenic mice enhanced hepatocyte proliferation after 2/3 partial hepatectomy (2/3 PHx). CR-1 over-expression significantly increased in vivo xenograft tumor growth of HCC cells in nude mice and in vitro HCC cell proliferation, migration, and invasion. CR-1 over-expression in the transgenic mouse livers deregulated HCC-related signaling pathways such as AKT, Wnt/β-catenin, Stat3, MAPK/ERK, JNK, TGF-β and Notch, as well as expression of HCC-related genes such as CD5L, S100A8, S100A9, Timd4, Orm2, Orm3, PDK4, DMBT1, G0S2, Plk2, Plk3, Gsta1 and Gsta2. However, histological signs of precancerous lesions, hepatocyte dysplasia or HCC formation were not observed in the livers of 3-, 6- or 8-month-old hepatocyte-specific CR-1-overexpressing transgenic mice. These findings demonstrate that liver-specific CR-1 overexpression in transgenic mice deregulates signaling pathways and genes associated with HCC.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Yan-Qing Li
- Department of Hematology, Central Hospital of Xuhui District, Shanghai 200030, China
| | - Shi-Hao Huang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yong-Long Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Jia-Wei Xia
- The Third People’s Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming 650041, China
| | - Jun-Shuang Jia
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Fang Wei
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Jia-Hong Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Guan-Qi Dai
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Yu-Cai Wang
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Yan Li
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Liu-Xin Han
- The Third People’s Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming 650041, China
| | - Xiao-Ling Zhang
- Department of Physiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541004, China
| | - Xu-Dong Xiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming 650118, China
| | - Wen-Tao Zhao
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming 650118, China
| | - Dong Xiao
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
- Institute of Comparative Medicine and Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Lin Lin
- Cancer Research Institute, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
9
|
Bremm JM, Boquett JA, Silva Michels M, Kowalski TW, Gomes FG, Vianna FSL, Vieira Sanseverino MT, Fraga LR. Investigating the role of EGF-CFC gene family in recurrent pregnancy loss through bioinformatics and molecular approaches. Syst Biol Reprod Med 2021; 67:450-462. [PMID: 34498535 DOI: 10.1080/19396368.2021.1965673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Recurrent pregnancy loss (RPL) is the most common reproductive failure, reaching 1-5% of women throughout their lives, and having unknown etiology in 50% of the cases. In humans, EGF-CFC1 (Epidermal Growth Factors & Cripto/FRL-1/Cryptic) gene family is composed by TDGF1 and CFC1, two developmental genes. The aim of this study was to investigate the role of EGF-CFC on RPL. To this, multiple approaches were performed; we conducted an expression analysis of TDGF1 and CFC1 using publicly available data from Gene Omnibus Expression (GEO), systems biology analyses and functional prediction; and a molecular analysis carried out in a case-control study. Our GEO analysis showed a decrease in TDGF1 expression in the endometrium (p=0.049) and CFC1 expression in placenta (p=0.015) of women with RPL. Network analysis, gene ontology and literature pointed to a strong connection between EGF-CFC1 gene family to pathways that play key roles during pregnancy, including TGF-β, c-Src/MAPK/AKT, Notch, TNFα, IFNγ and IL-6. A pathogenicity score developed for this gene family showed that the c.-14+1429T>C (rs3806702) variant in the TDGF1 and the p.Arg47Gln (rs201431919) variant in CFC1 gene would be the ones with the highest deleterious effect for RPL. In the case-control study, which involved 149 women with RPL and 159 controls, no statistical difference was observed in the allele and genotype distributions of the variants studied in the two groups. In this study, we performed extensive bioinformatics analysis for biomarker prioritization followed by experimental validation of proposed selected markers. Although there is no statistical difference in the frequencies of these variants between RPL and controls, the expression analysis results suggest that TDGF1 and CFC1 genes might play a role in RPL. In addition, systems biology analyzes raise the hypothesis that genes in other signaling pathways that may be related to RPL as good candidates for future studies.Abbreviations RPL: recurrent pregnancy loss; EGF-CFC1: Epidermal Growth Factors - Cripto/FRL-1; GEO: Gene Omnibus Expression; KEGG: Kyoto Encyclopedia of Genes and Genomes.
Collapse
Affiliation(s)
- João Matheus Bremm
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliano André Boquett
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Post-graduate Program in Child and Adolescent Health, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcus Silva Michels
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thayne Woycinck Kowalski
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Flávia Gobetti Gomes
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Post-Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Maria Teresa Vieira Sanseverino
- Post-graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,School of Medicine, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- Post-Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Morphological Science, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
10
|
Stifter K, Krieger J, Ruths L, Gout J, Mulaw M, Lechel A, Kleger A, Seufferlein T, Wagner M, Schirmbeck R. IFN-γ treatment protocol for MHC-I lo/PD-L1 + pancreatic tumor cells selectively restores their TAP-mediated presentation competence and CD8 T-cell priming potential. J Immunother Cancer 2021; 8:jitc-2020-000692. [PMID: 32868392 PMCID: PMC7462314 DOI: 10.1136/jitc-2020-000692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
Background Many cancer cells express a major histocompatibility complex class I low/ programmed cell death 1 ligand 1 positive (MHC-Ilo/PD-L1+) cell surface profile. For immunotherapy, there is, thus, an urgent need to restore presentation competence of cancer cells with defects in MHC-I processing/presentation combined with immune interventions that tackle the tumor-initiated PD-L1/PD-1 signaling axis. Using pancreatic ductal adenocarcinoma cells (PDACCs) as a model, we here explored if (and how) expression/processing of tumor antigens via transporters associated with antigen processing (TAP) affects priming of CD8 T cells in PD-1/PD-L1-competent/-deficient mice. Methods We generated tumor antigen-expressing vectors, immunized TAP-competent/-deficient mice and determined de novo primed CD8 T-cell frequencies by flow cytometry. Similarly, we explored the antigenicity and PD-L1/PD-1 sensitivity of PDACCs versus interferon-γ (IFN-γ)-treated PDACCs in PD-1/PD-L1-competent/deficient mice. The IFN-γ-induced effects on gene and cell surface expression profiles were determined by microarrays and flow cytometry. Results We identified two antigens (cripto-1 and an endogenous leukemia virus-derived gp70) that were expressed in the Endoplasmic Reticulum (ER) of PDACCs and induced CD8 T-cell responses either independent (Cripto-1:Kb/Cr16-24) or dependent (gp70:Kb/p15E) on TAP by DNA immunization. IFN-γ-treatment of PDACCs in vitro upregulated MHC-I- and TAP- but also PD-L1-expression. Mechanistically, PD-L1/PD-1 signaling was superior to the reconstitution of MHC-I presentation competence, as subcutaneously transplanted IFN-γ-treated PDACCs developed tumors in C57BL/6J and PD-L1-/- but not in PD-1-/- mice. Using PDACCs, irradiated at day 3 post-IFN-γ-treatment or PD-L1 knockout PDACCs as vaccines, we could selectively bypass upregulation of PD-L1, preferentially induce TAP-dependent gp70:Kb/p15E-specific CD8 T cells associated with a weakened PD-1+ exhaustion phenotype and reject consecutively injected tumor transplants in C57BL/6J mice. Conclusions The IFN-γ-treatment protocol is attractive for cell-based immunotherapies, because it restores TAP-dependent antigen processing in cancer cells, facilitates priming of TAP-dependent effector CD8 T-cell responses without additional check point inhibitors and could be combined with genetic vaccines that complement priming of TAP-independent CD8 T cells.
Collapse
Affiliation(s)
- Katja Stifter
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Jana Krieger
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Leonie Ruths
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Johann Gout
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, University Hospital Ulm, Ulm, Germany
| | - Andre Lechel
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | | | - Martin Wagner
- Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | |
Collapse
|
11
|
Arnouk H, Yum G, Shah D. Cripto-1 as a Key Factor in Tumor Progression, Epithelial to Mesenchymal Transition and Cancer Stem Cells. Int J Mol Sci 2021; 22:ijms22179280. [PMID: 34502188 PMCID: PMC8430685 DOI: 10.3390/ijms22179280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Cripto-1 is an essential protein for human development that plays a key role in the early phase of gastrulation in the differentiation of an embryo as well as assists with wound healing processes. Importantly, Cripto-1 induces epithelial to mesenchymal transition to turn fixed epithelial cells into a more mobile mesenchymal phenotype through the downregulation of epithelial adhesion molecules such as E-cadherin, occludins, and claudins, and the upregulation of mesenchymal, mobile proteins, such as N-cadherin, Snail, and Slug. Consequently, Cripto-1’s role in inducing EMT to promote cell motility is beneficial in embryogenesis, but detrimental in the formation, progression and metastasis of malignant tumors. Indeed, Cripto-1 is found to be upregulated in most cancers, such as breast, lung, gastrointestinal, hepatic, renal, cervical, ovarian, prostate, and skin cancers. Through its role in EMT, Cripto-1 can remodel cancer cells to enable them to travel through the extracellular matrix as well as blood and lymphatic vessels to metastasize to different organs. Additionally, Cripto-1 promotes the survival of cancer stem cells, which can lead to relapse in cancer patients.
Collapse
Affiliation(s)
- Hilal Arnouk
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
- Chicago College of Optometry, Midwestern University, Downers Grove, IL 60515, USA;
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA;
- College of Dental Medicine-Illinois, Midwestern University, Downers Grove, IL 60515, USA
- Correspondence:
| | - Gloria Yum
- Chicago College of Optometry, Midwestern University, Downers Grove, IL 60515, USA;
| | - Dean Shah
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA;
- Master of Public Health Program, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
12
|
Ishii H, Zahra MH, Takayanagi A, Seno M. A Novel Artificially Humanized Anti-Cripto-1 Antibody Suppressing Cancer Cell Growth. Int J Mol Sci 2021; 22:ijms22041709. [PMID: 33567764 PMCID: PMC7915030 DOI: 10.3390/ijms22041709] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Cripto-1 is a member of the EGF-CFC/FRL1/Cryptic family and is involved in embryonic development and carcinogenesis. We designed a novel anti-Cripto-1 artificial antibody and assessed the recognition to the antigen and the potential to suppress the growth of cancer stem cells. First, single chain antibody clones were isolated by bio-panning with the affinity to recombinant Cripto-1 protein from our original phage-display library. Then, the variable regions of heavy chain VH and light chain VL in each clone were fused to constant regions of heavy chain CH and light chain CL regions respectively. These fused genes were expressed in ExpiCHO-S cells to produce artificial humanized antibodies against Cripto-1. After evaluation of the expression levels, one clone was selected and the anti-Cripto-1 antibody was produced and purified. The purified antibody showed affinity to recombinant Cripto-1 at 1.1 pmol and immunoreactivity to cancer tissues and cell lines. The antibody was available to detect the immunoreactivity in tissue microarrays of malignant tumors as well as in Cripto-1 overexpressing cells. Simultaneously, the antibody exhibited the potential to suppress the growth of human colon cancer derived GEO cells overexpressing Cripto-1 with IC50 at approximately 110 nM. The artificially humanized antibody is proposed to be a good candidate to target cancer cells overexpressing Cripto-1.
Collapse
Affiliation(s)
- Hiroko Ishii
- GSP Enterprise, Inc., 1-4-38 12F Minato-machi, Naniwaku, Osaka 556-0017, Japan; (H.I.); (A.T.)
| | - Maram H. Zahra
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan;
| | - Atushi Takayanagi
- GSP Enterprise, Inc., 1-4-38 12F Minato-machi, Naniwaku, Osaka 556-0017, Japan; (H.I.); (A.T.)
| | - Masaharu Seno
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan;
- Correspondence: ; Tel./Fax: +81-86-251-8216
| |
Collapse
|
13
|
Silva AF, Silva ECCDA, Oliveira CN, Rocha KBF, Miguel MCC, Pereira JS. Oral and uterine leiomyomas exhibit high immunoexpression of Cripto-1 compared to normal myometrium. AN ACAD BRAS CIENC 2020; 92:e20200519. [PMID: 33331446 DOI: 10.1590/0001-3765202020200519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/21/2020] [Indexed: 11/22/2022] Open
Abstract
Leiomyomas are the most common benign tumors in women. Many of them are associated with significant morbidity. The present study aimed to analyze histomorphological and histochemical characteristics and immunoexpression of Cripto-1 in oral leiomyomas (OL), uterine leiomyomas (UL), and normal myometrium (NM). Sample was composed of ten OL, 11 UL and 11 NM. Histomorphological characteristics were analyzed at 100 and 400x magnifications with HE staining. The immunoexpression of Cripto-1 was analyzed in five high-power fields. Statistical analysis considered a significant difference when p<0.05. Six OL disclosed moderate/intense inflammatory infiltrate, while ten UL exhibited mild infiltrate (p=0.024). When analyzing all leiomyomas together, 20 exhibited hyalinization, whereas no NM exhibited this alteration (p<0.001). There was no statistical difference in the distribution of mast cells among the lesions. The median Cripto-1 was higher in UL (9.0), followed by OL and NM (4.0). Associations of the Cripto-1 expression between leiomyomas (separately and together) and NM were statistically significant (p<0.001). These results indicate that OL and UL exhibit similar histomorphological and histochemical characteristics, as well as differences to NM. The higher immunoexpression of Cripto-1 in leiomyomas compared to NM suggests that this protein may influence cell proliferation and tissue architecture of oral and uterine leiomyomas.
Collapse
Affiliation(s)
- AndrÉ F Silva
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Emmily C C DA Silva
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - ClÁudia N Oliveira
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Keyla B F Rocha
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Marcia C C Miguel
- Programa de Pós-Graduação em Patologia Oral, Universidade Federal do Rio Grande do Norte, Rua Senador Salgado Filho, 1787, Lagoa Nova, 59056-000 Natal, RN, Brazil
| | - Joabe S Pereira
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| |
Collapse
|
14
|
Raos D, Krasic J, Masic S, Abramovic I, Coric M, Kruslin B, Katusic Bojanac A, Bulic-Jakus F, Jezek D, Ulamec M, Sincic N. In Search of TGCT Biomarkers: A Comprehensive In Silico and Histopathological Analysis. DISEASE MARKERS 2020; 2020:8841880. [PMID: 33224314 PMCID: PMC7666710 DOI: 10.1155/2020/8841880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/10/2020] [Accepted: 07/31/2020] [Indexed: 01/22/2023]
Abstract
Testicular germ cell tumors (TGCTs) are ever more affecting the young male population. Germ cell neoplasia in situ (GCNIS) is the origin of TGCTs, namely, seminomas (SE) and a heterogeneous group of nonseminomas (NS) comprising embryonal carcinoma, teratoma, yolk sac tumor, and choriocarcinoma. Response to the treatment and prognosis, especially of NS, depend on precise diagnosis with a necessity for discovery of new biomarkers. We aimed to perform comprehensive in silico analysis at the DNA, RNA, and protein levels of six prospective (HOXA9, MGMT, CFC1, PRSS21, RASSF1A, and MAGEC2) and six known TGCT biomarkers (OCT4, SOX17, SOX2, SALL4, NANOG, and KIT) and assess its congruence with histopathological analysis in all forms of TGCTs. Cancer Hallmarks Analytics Tool, the Search Tool for the Retrieval of Interacting Genes/Proteins database, and UALCAN, an interactive web resource for analyzing cancer OMICS data, were used. In 108 TGCT and 48 tumor-free testicular samples, the immunoreactivity score (IRS) was calculated. SE showed higher frequency in DNA alteration, while DNA methylation was significantly higher for all prospective biomarkers in NS. In GCNIS, we assessed the clinical positivity of RASSF1 and PRSS21 in 52% and 62% of samples, respectively, in contrast to low or nil positivity in healthy seminiferous tubules, TGTCs as a group, SE, NS, or all NS components. Although present in approximately 80% of healthy seminiferous tubules (HT) and GCNIS, HOXA9 was diagnostically positive in 64% of TGCTs, while it was positive in 82% of NS versus 29% of SE. Results at the DNA, mRNA, and protein levels on putative and already known biomarkers were included in the suggested panels that may prove to be important for better diagnostics of various forms of TGCTs.
Collapse
Affiliation(s)
- Dora Raos
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Jure Krasic
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Silvija Masic
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre Milosrdnice University Hospital Center, Vinogradska Cesta 29, 10000 Zagreb, Croatia
| | - Irena Abramovic
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Marijana Coric
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Department of Pathology and Cytology, University Hospital Centre Zagreb, Kišpatićeva Ulica 12, 10000 Zagreb, Croatia
| | - Bozo Kruslin
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre Milosrdnice University Hospital Center, Vinogradska Cesta 29, 10000 Zagreb, Croatia
| | - Ana Katusic Bojanac
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Floriana Bulic-Jakus
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Davor Jezek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| | - Monika Ulamec
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Ljudevit Jurak Clinical Department of Pathology and Cytology, Sestre Milosrdnice University Hospital Center, Vinogradska Cesta 29, 10000 Zagreb, Croatia
- Department of Pathology, University of Zagreb School of Dental Medicine and School of Medicine, Gundulićeva Ulica 5, 10000 Zagreb, Croatia
| | - Nino Sincic
- Department of Medical Biology, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Group for Research on Epigenetic Biomarkers, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Šalata 3, 10000 Zagreb, Croatia
| |
Collapse
|
15
|
Ninkovic S, Harrison SJ, Quach H. Glucose-regulated protein 78 (GRP78) as a potential novel biomarker and therapeutic target in multiple myeloma. Expert Rev Hematol 2020; 13:1201-1210. [PMID: 32990063 DOI: 10.1080/17474086.2020.1830372] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Glucose-regulated protein 78 (GRP78) is a stress-inducible molecular chaperone expressed within the endoplasmic reticulum where it acts as a master regulator of the unfolded protein response (UPR) pathway. At times of ER stress, activation of the UPR, a multimolecular pathway, limits proteotoxicity induced by misfolded proteins. In malignancies, including multiple myeloma which is characterized by an accumulation of misfolded immunoglobulins, GRP78 expression is increased, with notable translocation of GRP78 to the cell surface. Studies suggest cell-surface GRP78 (csGRP78) to be of prognostic significance with emerging evidence that it interacts with a myriad of co-ligands to activate signaling pathways promoting cell proliferation and survival or apoptosis. AREAS COVERED This review focuses on the role of ER and csGRP78 in physiology and oncogenesis in multiple myeloma, addressing factors that shift the balance in GRP78 signaling from survival to apoptosis. The role of GRP78 as a potential prognostic biomarker is explored and current therapeutics in development aimed at targeting csGRP78 are addressed. We conducted a PubMed literature search using the keywords 'GRP78,' 'multiple myeloma' reviewing studies prior to 2020. EXPERT OPINION Cell-surface GRP78 expression is a potential novel prognostic biomarker in myeloma and targeting of csGRP78 is promising and requires further investigation.
Collapse
Affiliation(s)
- Slavisa Ninkovic
- Department of Haematology, St. Vincent's Hospital Melbourne , Fitzroy, Australia.,Department of Medicine, University of Melbourne , Fitzroy, Australia
| | - Simon J Harrison
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital , Melbourne, Australia.,Sir Peter MacCallum Dept of Oncology, University of Melbourne , Parkville, Australia
| | - Hang Quach
- Department of Haematology, St. Vincent's Hospital Melbourne , Fitzroy, Australia.,Department of Medicine, University of Melbourne , Fitzroy, Australia
| |
Collapse
|
16
|
ER Stress-Induced Secretion of Proteins and Their Extracellular Functions in the Heart. Cells 2020; 9:cells9092066. [PMID: 32927693 PMCID: PMC7563782 DOI: 10.3390/cells9092066] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a result of conditions that imbalance protein homeostasis or proteostasis at the ER, for example ischemia, and is a common event in various human pathologies, including the diseased heart. Cardiac integrity and function depend on the active secretion of mature proteins from a variety of cell types in the heart, a process that requires an intact ER environment for efficient protein folding and trafficking to the secretory pathway. As a consequence of ER stress, most protein secretion by the ER secretory pathway is decreased. Strikingly, there is a select group of proteins that are secreted in greater quantities during ER stress. ER stress resulting from the dysregulation of ER Ca2+ levels, for instance, stimulates the secretion of Ca2+-binding ER chaperones, especially GRP78, GRP94, calreticulin, and mesencephalic astrocyte-derived neurotrophic factor (MANF), which play a multitude of roles outside the cell, strongly depending on the cell type and tissue. Here we review current insights in ER stress-induced secretion of proteins, particularly from the heart, and highlight the extracellular functions of these proteins, ranging from the augmentation of cardiac cell viability to the modulation of pro- and anti-apoptotic, oncogenic, and immune-stimulatory cell signaling, cell invasion, extracellular proteostasis, and more. Many of the roles of ER stress-induced protein secretion remain to be explored in the heart. This article is part of a special issue entitled “The Role of Proteostasis Derailment in Cardiac Diseases.”
Collapse
|
17
|
Rodrigues Sousa E, Zoni E, Karkampouna S, La Manna F, Gray PC, De Menna M, Kruithof-de Julio M. A Multidisciplinary Review of the Roles of Cripto in the Scientific Literature Through a Bibliometric Analysis of its Biological Roles. Cancers (Basel) 2020; 12:cancers12061480. [PMID: 32517087 PMCID: PMC7352664 DOI: 10.3390/cancers12061480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Cripto is a small glycosylphosphatidylinisitol (GPI)-anchored and secreted oncofetal protein that plays important roles in regulating normal physiological processes, including stem cell differentiation, embryonal development, and tissue growth and remodeling, as well as pathological processes such as tumor initiation and progression. Cripto functions as a co-receptor for TGF-β ligands such as Nodal, GDF1, and GDF3. Soluble and secreted forms of Cripto also exhibit growth factor-like activity and activate SRC/MAPK/PI3K/AKT pathways. Glucose-Regulated Protein 78 kDa (GRP78) binds Cripto at the cell surface and has been shown to be required for Cripto signaling via both TGF-β and SRC/MAPK/PI3K/AKT pathways. To provide a comprehensive overview of the scientific literature related to Cripto, we performed, for the first time, a bibliometric analysis of the biological roles of Cripto as reported in the scientific literature covering the last 10 years. We present different fields of knowledge in comprehensive areas of research on Cripto, ranging from basic to translational research, using a keyword-driven approach. Our ultimate aim is to aid the scientific community in conducting targeted research by identifying areas where research has been conducted so far and, perhaps more importantly, where critical knowledge is still missing.
Collapse
Affiliation(s)
- Elisa Rodrigues Sousa
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Eugenio Zoni
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Sofia Karkampouna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Federico La Manna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Marta De Menna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Marianna Kruithof-de Julio
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
18
|
Igder S, Mohammadiasl J, Azadpour S, Mansouri E, Ashktorab H, Mokarram P. KRAS mutation and abnormal expression of Cripto-1 as two potential candidate biomarkers for detection of colorectal cancer development. J Cell Biochem 2019; 121:2901-2908. [PMID: 31692030 DOI: 10.1002/jcb.29526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/10/2019] [Indexed: 12/31/2022]
Abstract
Colorectal cancer (CRC), regardless of standard procedures of treatment and screening, is still considered one of the deadliest cancers in the Western world, and in economically developed Asian countries, especially Iran. The current study was undertaken to investigate whether changes in the level of Cripto-1 (CR-1) expression and KRAS mutations have a cumulative effect on the onset and progression of CRC. Fifty colorectal tissue samples, including 35 colorectal carcinomas with matching adjacent mucosa, and 15 colorectal adenomas, were chosen for analysis. Twenty-five CRC biopsies and 15 adenoma were analyzed for KRAS mutations by DNA sequencing (Sanger sequencing), and all 50 patients (35 CRCs and 15 adenomas) were evaluated by immunohistochemistry for the CR-1 protein expression. The inducible somatic KRAS mutation (G12D) was observed in nine (36%) of CRC patients, and in two (13.3%) of adenoma patients. The CR-1 expression level in both adenomas (P < .05) and carcinomas (P < .001), were significantly different, compared with the matching adjacent mucosa. The intensity of CR-1 staining in adenomas was less than the intensity of staining, detected in the CRCs (P < .001). The G12D KRAS mutation and CR-1 abnormalities are significantly associated as two signature biomarkers with potential clinical characteristics for the detection of CRC development.
Collapse
Affiliation(s)
- Somayeh Igder
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Mohammadiasl
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shima Azadpour
- Faculty Member of Hematology Department, Abadan School of Medical Sciences, Abadan, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Ashktorab
- Department of Medicine and Cancer Center, Howard University College of Medicine, Washington, District of Columbia
| | - Pooneh Mokarram
- Colorectal Cancer Research Center, Department of Biochemistry, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
20
|
Alowaidi F, Hashimi SM, Alqurashi N, Wood SA, Wei MQ. Cripto-1 overexpression in U87 glioblastoma cells activates MAPK, focal adhesion and ErbB pathways. Oncol Lett 2019; 18:3399-3406. [PMID: 31452820 DOI: 10.3892/ol.2019.10626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022] Open
Abstract
Discovering the underlying signalling pathways that control cancer cells is crucial for understanding their biology and to develop therapeutic regimens. Thus, the aim of the present study was to determine the effect of Cripto-1 on pathways controlling glioblastoma (GBM) cell function. To this end, changes in protein phosphorylation in cells overexpressing Cripto-1 were analysed using the Kyoto Encyclopedia of Genes and Genomes pathway analysis tool, as well as the Uniprot resource to identify the functions of Cripto-1-dependent phosphorylated proteins. This revealed that proteins affected by Cripto-1 overexpression are involved in multiple signalling pathways. The mitogen-activated protein kinase (MAPK), focal adhesion (FA) and ErbB pathways were found to be enriched by Cripto-1 overexpression with 35, 27 and 24% of pathway proteins phosphorylated, respectively. These pathways control important cellular processes in cancer cells that correlate with the observed functional changes described in earlier studies. More specifically, Cripto-1 may regulate MAPK cellular proliferation and survival pathways by activating epithelial growth factor receptor (EGFR; Ser1070) or fibroblast GFR1 (Tyr654). Its effect on cellular proliferation and survival could be mediated through Src (Tyr418), FA kinase (FAK; Tyr396), p130CAS (Tyr410), c-Jun (Ser63), Paxillin (PXN; Tyr118) and BCL2 (Thr69) of the FA pathway. Cripto-1 may also control cellular motility and invasion by activating Src (Tyr418), FAK (Tyr396) and PXN (Tyr118) of the FA pathway. However, Cripto-1 regulation of cellular invasion and migration might be not limited to the FA pathway, it may also control these cellular mechanisms through signalling via EGFR (Ser1070)/Her2 (Tyr877) to mediate the Src (Tyr418) and FAK (Tyr396) cascade activation of the ErbB signalling pathway. Angiogenesis could be mediated by Cripto-1 by activating c-Jun (Ser63) through EGFR (Ser1070)/Her2 (Tyr877) of the ErbB pathway. To conclude, the present study has augmented and enriched our current knowledge on the crucial roles that Cripto-1 may play in controlling different cellular mechanisms in GBM cells.
Collapse
Affiliation(s)
- Faisal Alowaidi
- Department of Pathology and Laboratory Medicine, College of Medicine and University Hospital, King Saud University, Riyadh 11461, Saudi Arabia
| | - Saeed M Hashimi
- Department of Basic Science, Biology Unit, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Naif Alqurashi
- Department of Basic Science, Biology Unit, Deanship of Preparatory Year and Supporting Studies, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Stephen A Wood
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Ming Q Wei
- Division of Molecular and Gene Therapies, School of Medical Science, Griffith University, Gold Coast, Queensland 4222, Australia
| |
Collapse
|
21
|
Liu Y, Wang J, Yang T, Liu R, Xu Y. Overexpression levels of cripto-1 predict poor prognosis in patients with prostate cancer following radical prostatectomy. Oncol Lett 2019; 18:2584-2591. [PMID: 31452743 DOI: 10.3892/ol.2019.10555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 05/17/2019] [Indexed: 12/28/2022] Open
Abstract
Overexpression of cripto-1 (CR-1), an epidermal growth factor-cripto-1/FRL-1/Cryptic family protein, has been reported in multiple types of malignancy. However, the clinical functions of CR-1 in prostate cancer (PCa) remain largely unclear. The objective of the present study was to investigate the association between CR-1 expression and the clinicopathological features and prognosis of PCa. CR-1 expression was evaluated in 138 PCa tissues and 67 benign prostate hyperplasia (BPH) tissues using immunohistochemistry. The association between the clinicopathological features of patients with PCa and CR-1 expression was analyzed using a χ2 test. Receiver operating characteristic (ROC) curve and Cox regression model were used to analyze the association between CR-1 expression and biochemical recurrence (BCR)-free survival. It was revealed that the protein expression of CR-1 was markedly higher in PCa tissues than in BPH tissues. The mRNA expression of CR-1 in PCa tissue and cells was also significantly higher than in BPH tissue and the normal RWPE-1 prostate cell line (P<0.05). In addition, high CR-1 expression was significantly associated with prostate-specific antigen level (P=0.008), Gleason score (P=0.011) and lymph node metastasis (P=0.025) in patients with PCa. ROC curve indicated that patients with elevated expression of CR-1 exhibited shorter BCR-free survival (P<0.001). Furthermore, multivariate statistical analysis demonstrated that overexpression of CR-1 may be a novel predictor for prognosis of patients with PCa. Accordingly, the present study considered CR-1 to be a valuable predictor of poor prognosis and progression in PCa, and a potential therapeutic target for patients with PCa.
Collapse
Affiliation(s)
- Yan Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jianan Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Tong Yang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Ranlu Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yong Xu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China.,Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
22
|
Alowaidi F, Hashimi SM, Nguyen M, Meshram M, Alqurashi N, Cavanagh BL, Bellette B, Ivanovski S, Meedenyia A, Wood SA. Investigating the role of CRIPTO-1 (TDGF-1) in glioblastoma multiforme U87 cell line. J Cell Biochem 2019; 120:7412-7427. [PMID: 30426531 DOI: 10.1002/jcb.28015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 10/10/2018] [Indexed: 01/24/2023]
Abstract
Cripto-1 has been implicated in a number of human cancers. Although there is high potential for a role of Cripto-1 in glioblastoma multiforme (GBM) pathogenesis and progression, few studies have tried to define its role in GBM. These studies were limited in that Cripto-1 expression was not studied in detail in relation to markers of cancer initiation and progression. Therefore, these correlative studies allowed limited interpretation of Criptos-1's effect on the various aspects of GBM development using the U87 GBM cell line. In this study, we sought to delineate the role of Cripto-1 in facilitating pathogenesis, stemness, proliferation, invasion, migration and angiogenesis in GBM. Our findings show that upon overexpressing Cripto-1 in U87 GBM cells, the stemness markers Nanog, Oct4, Sox2, and CD44 increased expression. Similarly, an increase in Ki67 was observed demonstrating Cripto-1's potential to induce cellular proliferation. Likewise, we report a novel finding that increased expression of the markers of migration and invasion, Vimentin and Twist, correlated with upregulation of Cripto-1. Moreover, Cripto-1 exposure led to VEGFR-2 overexpression along with higher tube formation under conditions promoting endothelial growth. Taken together our results support a role for Cripto-1 in the initiation, development, progression, and maintenance of GBM pathogenesis. The data presented here are also consistent with a role for Cripto-1 in the re-growth and invasive growth in GBM. This highlights its potential use as a predictive and diagnostic marker in GBM as well as a therapeutic target.
Collapse
Affiliation(s)
- Faisal Alowaidi
- Department of Pathology and Laboratory Medicine, College of Medicine and University Hospitals, King Saud University, Riyadh, Saudi Arabia.,Menzies Health Institute Queensland, School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Saeed M Hashimi
- Department of Basic Science, Biology Unit, Deanship of Preparatory Year and Supporting studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Maria Nguyen
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mallika Meshram
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Naif Alqurashi
- Department of Basic Science, Biology Unit, Deanship of Preparatory Year and Supporting studies, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Brenton L Cavanagh
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Bernadette Bellette
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Saso Ivanovski
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Adrian Meedenyia
- Menzies Health Institute Queensland, School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Stephen A Wood
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| |
Collapse
|
23
|
Cultrara CN, Kozuch SD, Ramasundaram P, Heller CJ, Shah S, Beck AE, Sabatino D, Zilberberg J. GRP78 modulates cell adhesion markers in prostate Cancer and multiple myeloma cell lines. BMC Cancer 2018; 18:1263. [PMID: 30563499 PMCID: PMC6299583 DOI: 10.1186/s12885-018-5178-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/04/2018] [Indexed: 12/22/2022] Open
Abstract
Background Glucose regulated protein 78 (GRP78) is a resident chaperone of the endoplasmic reticulum and a master regulator of the unfolded protein response under physiological and pathological cell stress conditions. GRP78 is overexpressed in many cancers, regulating a variety of signaling pathways associated with tumor initiation, proliferation, adhesion and invasion which contributes to metastatic spread. GRP78 can also regulate cell survival and apoptotic pathways to alter responsiveness to anticancer drugs. Tumors that reside in or metastasize to the bone and bone marrow (BM) space can develop pro-survival signals through their direct adhesive interactions with stromal elements of this niche thereby resisting the cytotoxic effects of drug treatment. In this study, we report a direct correlation between GRP78 and the adhesion molecule N-cadherin (N-cad), known to play a critical role in the adhesive interactions of multiple myeloma and metastatic prostate cancer with the bone microenvironment. Methods N-cad expression levels (transcription and protein) were evaluated upon siRNA mediated silencing of GRP78 in the MM.1S multiple myeloma and the PC3 metastatic prostate cancer cell lines. Furthermore, we evaluated the effects of GRP78 knockdown (KD) on epithelial-mesenchymal (EMT) transition markers, morphological changes and adhesion of PC3 cells. Results GRP78 KD led to concomitant downregulation of N-cad in both tumors types. In PC3 cells, GRP78 KD significantly decreased E-cadherin (E-cad) expression likely associated with the induction in TGF-β1 expression. Furthermore, GRP78 KD also triggered drastic changes in PC3 cells morphology and decreased their adhesion to osteoblasts (OSB) dependent, in part, to the reduced N-cad expression. Conclusion This work implicates GRP78 as a modulator of cell adhesion markers in MM and PCa. Our results may have clinical implications underscoring GRP78 as a potential therapeutic target to reduce the adhesive nature of metastatic tumors to the bone niche. Electronic supplementary material The online version of this article (10.1186/s12885-018-5178-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher N Cultrara
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Stephen D Kozuch
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Poornema Ramasundaram
- Center for Discovery and Innovation, Hackensack University Medical Center, 340 Kingsland Street, Building 102, Nutley, NJ, 07110, USA
| | - Claudia J Heller
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Sunil Shah
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Adah E Beck
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - David Sabatino
- Department of Chemistry and Biochemistry, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Jenny Zilberberg
- Center for Discovery and Innovation, Hackensack University Medical Center, 340 Kingsland Street, Building 102, Nutley, NJ, 07110, USA.
| |
Collapse
|
24
|
Müller GA. The release of glycosylphosphatidylinositol-anchored proteins from the cell surface. Arch Biochem Biophys 2018; 656:1-18. [DOI: 10.1016/j.abb.2018.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
|
25
|
Xu CH, Chi CZ, Zhang Q, Wang YC, Wang W, Yuan Q, Zhan P, Zhang XW, Lin Y. Diagnostic and prognostic value of serum Cripto-1 in patients with non-small cell lung cancer. CLINICAL RESPIRATORY JOURNAL 2018; 12:2469-2474. [PMID: 29570945 DOI: 10.1111/crj.12793] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/12/2018] [Accepted: 03/11/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Chun Hua Xu
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Chuan Zhen Chi
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Qian Zhang
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Yu Chao Wang
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Wei Wang
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Qi Yuan
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Ping Zhan
- Endoscopic Center of Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
- Clinical Center of Nanjing Respiratory Diseases and Imaging; Nanjing Jiangsu 210029 People's Republic of China
| | - Xiu Wei Zhang
- Department of Respiratory Medicine; Nanjing Jiangning Hospital; Nanjing Jiangsu 211100 People's Republic of China
| | - Yong Lin
- Department of Respiratory Medicine; Nanjing Chest Hospital; Nanjing Jiangsu 210029 People's Republic of China
| |
Collapse
|
26
|
Zhang Y, Xu H, Chi X, Fan Y, Shi Y, Niu J. High level of serum Cripto-1 in hepatocellular carcinoma, especially with hepatitis B virus infection. Medicine (Baltimore) 2018; 97:e11781. [PMID: 30170372 PMCID: PMC6392992 DOI: 10.1097/md.0000000000011781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Human Cripto-1 (CR-1), a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic protein family (EGF-CFC), is highly expressed in a variety of human cancers. We aimed to detect serum CR-1 level in liver diseases especially in hepatocellular carcinoma (HCC) patients. METHODS Serum CR-1 level was Sandwich-type enzyme-linked immuno sorbent assay (ELISA) detected in 330 patients with liver diseases including HCC, cirrhosis, and chronic hepatitis and 50 volunteers without hepatitis B virus (HBV) or hepatitis C virus (HCV) infection as control. RESULTS The serum CR-1 level was significantly higher in HCC patients than volunteer controls and it was also significantly higher in HBV-related HCC than HCV-related HCC. In addition, serum CR-1 level was correlated with serum alpha-feto-protein (AFP) in HBV-related HCC patients. The serum CR-1 was also higher in cirrhosis and chronic hepatitis than volunteer controls. The serum CR-1 in HBV-related cirrhosis was higher than chronic hepatitis B, but there was no significant difference between HCV-related cirrhosis and chronic hepatitis C. CONCLUSIONS Serum CR-1 was higher in HCC patients and might serve as a complementary biomarker to clinical diagnosis of HBV-related HCC. The high level of serum CR-1 in HBV-related liver disease might be partly attributed to HBV infection.
Collapse
Affiliation(s)
- Yingyu Zhang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | | |
Collapse
|
27
|
Karkampouna S, van der Helm D, Gray PC, Chen L, Klima I, Grosjean J, Burgmans MC, Farina-Sarasqueta A, Snaar-Jagalska EB, Stroka DM, Terracciano L, van Hoek B, Schaapherder AF, Osanto S, Thalmann GN, Verspaget HW, Coenraad MJ, Kruithof-de Julio M. CRIPTO promotes an aggressive tumour phenotype and resistance to treatment in hepatocellular carcinoma. J Pathol 2018; 245:297-310. [PMID: 29604056 DOI: 10.1002/path.5083] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/13/2018] [Accepted: 03/26/2018] [Indexed: 12/25/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Despite increasing treatment options for this disease, prognosis remains poor. CRIPTO (TDGF1) protein is expressed at high levels in several human tumours and promotes oncogenic phenotype. Its expression has been correlated to poor prognosis in HCC. In this study, we aimed to elucidate the basis for the effects of CRIPTO in HCC. We investigated CRIPTO expression levels in three cohorts of clinical cirrhotic and HCC specimens. We addressed the role of CRIPTO in hepatic tumourigenesis using Cre-loxP-controlled lentiviral vectors expressing CRIPTO in cell line-derived xenografts. Responses to standard treatments (sorafenib, doxorubicin) were assessed directly on xenograft-derived ex vivo tumour slices. CRIPTO-overexpressing patient-derived xenografts were established and used for ex vivo drug response assays. The effects of sorafenib and doxorubicin treatment in combination with a CRIPTO pathway inhibitor were tested in ex vivo cultures of xenograft models and 3D cultures. CRIPTO protein was found highly expressed in human cirrhosis and hepatocellular carcinoma specimens but not in those of healthy participants. Stable overexpression of CRIPTO in human HepG2 cells caused epithelial-to-mesenchymal transition, increased expression of cancer stem cell markers, and enhanced cell proliferation and migration. HepG2-CRIPTO cells formed tumours when injected into immune-compromised mice, whereas HepG2 cells lacking stable CRIPTO overexpression did not. High-level CRIPTO expression in xenograft models was associated with resistance to sorafenib, which could be modulated using a CRIPTO pathway inhibitor in ex vivo tumour slices. Our data suggest that a subgroup of CRIPTO-expressing HCC patients may benefit from a combinatorial treatment scheme and that sorafenib resistance may be circumvented by inhibition of the CRIPTO pathway. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sofia Karkampouna
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland
| | - Danny van der Helm
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter C Gray
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Lanpeng Chen
- Institute of Biology, Department of Molecular Cell Biology, Leiden University, Leiden, The Netherlands
| | - Irena Klima
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland
| | - Joël Grosjean
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland
| | - Mark C Burgmans
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Ewa B Snaar-Jagalska
- Institute of Biology, Department of Molecular Cell Biology, Leiden University, Leiden, The Netherlands
| | - Deborah M Stroka
- Department of Biomedical Research, Visceral Surgery and Medicine, University of Bern, Bern University Hospital, Switzerland
| | - Luigi Terracciano
- Molecular Pathology Division, Institute of Pathology, University Hospital Basel, Switzerland
| | - Bart van Hoek
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Susan Osanto
- Department of Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - George N Thalmann
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland.,Department of Urology, Bern University Hospital, Switzerland
| | - Hein W Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Minneke J Coenraad
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marianna Kruithof-de Julio
- Department of Biomedical Research, Urology Group, University of Bern, Bern, Switzerland.,Department of Urology, Bern University Hospital, Switzerland
| |
Collapse
|
28
|
Alekseenko IV, Vinogradova TV, Sverdlov ED. Genetic Regulatory Mechanisms of Evolution and Embryogenesis in a Distorting Mirror of Carcinogenesis. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
29
|
Nickel J, Ten Dijke P, Mueller TD. TGF-β family co-receptor function and signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:12-36. [PMID: 29293886 DOI: 10.1093/abbs/gmx126] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-β (TGF-β) family members, which include TGF-βs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-β family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-β family members. While some appear to be specific to TGF-β family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-β family co-receptors, which continue to be discovered.
Collapse
Affiliation(s)
- Joachim Nickel
- Universitätsklinikum Würzburg, Lehrstuhl für Tissue Engineering und Regenerative Medizin und Fraunhofer Institut für Silicatforschung (ISC), Translationszentrum "Regenerative Therapien", Röntgenring 11, D-97070 Würzburg, Germany
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Thomas D Mueller
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von-Sachs Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs-Platz 2, D-97082 Würzburg, Germany
| |
Collapse
|
30
|
CRIPTO and its signaling partner GRP78 drive the metastatic phenotype in human osteotropic prostate cancer. Oncogene 2017; 36:4739-4749. [PMID: 28394345 PMCID: PMC5562855 DOI: 10.1038/onc.2017.87] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 02/10/2017] [Accepted: 02/26/2017] [Indexed: 12/21/2022]
Abstract
CRIPTO (CR-1, TDGF1) is a cell surface/secreted oncoprotein actively involved in development and cancer. Here, we report that high expression of CRIPTO correlates with poor survival in stratified risk groups of prostate cancer (PCa) patients. CRIPTO and its signaling partner glucose-regulated protein 78 (GRP78) are highly expressed in PCa metastases and display higher levels in the metastatic ALDHhigh sub-population of PC-3M-Pro4Luc2 PCa cells compared with non-metastatic ALDHlow. Coculture of the osteotropic PC-3M-Pro4Luc2 PCa cells with differentiated primary human osteoblasts induced CRIPTO and GRP78 expression in cancer cells and increases the size of the ALDHhigh sub-population. Additionally, CRIPTO or GRP78 knockdown decreases proliferation, migration, clonogenicity and the size of the metastasis-initiating ALDHhigh sub-population. CRIPTO knockdown reduces the invasion of PC-3M-Pro4Luc2 cells in zebrafish and inhibits bone metastasis in a preclinical mouse model. These results highlight a functional role for CRIPTO and GRP78 in PCa metastasis and suggest that targeting CRIPTO/GRP78 signaling may have significant therapeutic potential.
Collapse
|
31
|
Liu Y, Qin Z, Yang K, Liu R, Xu Y. Cripto-1 promotes epithelial-mesenchymal transition in prostate cancer via Wnt/β-catenin signaling. Oncol Rep 2017; 37:1521-1528. [PMID: 28098905 DOI: 10.3892/or.2017.5378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/21/2016] [Indexed: 11/05/2022] Open
Abstract
The Cripto-1 (CR-1) derived EGF-CFC family was overexpressed in tumor development enhancing proliferation, epithelial-mesenchymal transition (EMT) and migration of tumor cells. However, correlation between CR-1 and prostate cancer (PCa) remains still unclear. In the present study, we proved that CR-1 was expressed in PCa and its function was in the progression of PCa. Compared with benign prostatic hyperplasia (BPH) tissues, we confirmed that PCa tissues had high expression of CR-1 by immunohistochemistry and statistical data showed that CR-1 promoted properties of EMT in PCa tissues, including the downregulation of the cell adhesion molecules β-catenin (membrane) and E-cadherin while upregulating transcription factors β-catenin. Overexpression of CR-1 had close relationship with PSA, Gleason, clinical staging and lymph node metastasis in PCa patients. Then, we found that PC-3 cells transfected with CR-1-shRNA inhibited EMT using RT-PCR, RT-qPCR, western blotting and immunofluorescence. Also, we evaluated cell invasive ability in vitro by transwell and wound-healing assay. Our data showed that transfected CR-1-shRNA altered EMT including β-catenin, E-cadherin, c-myc, GSK-3, p-GSK and Wnt/β-catenin pathway in PC-3. It also suppressed PC-3 cell migration. Additionally, our results displayed that Licl had antitumor activity against PC-3 through the inhibition of Wnt/β-catenin pathway. Inhibition of cell viability was dose-time dependent. The present study proved that CR-1 regulates EMT of PCa by Wnt/β-catenin pathway. Hence, CR-1 may provide a new biological marker, and possibly contributes to clinical treatment against PCa.
Collapse
Affiliation(s)
- Yan Liu
- Prostate Disease Laboratory, Tianjin Institute of Urology, Tianjin 300211, P.R. China
| | - Zhenbang Qin
- Prostate Disease Laboratory, Tianjin Institute of Urology, Tianjin 300211, P.R. China
| | - Kuo Yang
- Prostate Disease Laboratory, Tianjin Institute of Urology, Tianjin 300211, P.R. China
| | - Ranlu Liu
- Prostate Disease Laboratory, Tianjin Institute of Urology, Tianjin 300211, P.R. China
| | - Yong Xu
- Prostate Disease Laboratory, Tianjin Institute of Urology, Tianjin 300211, P.R. China
| |
Collapse
|
32
|
Lee GH, Fujita M, Takaoka K, Murakami Y, Fujihara Y, Kanzawa N, Murakami KI, Kajikawa E, Takada Y, Saito K, Ikawa M, Hamada H, Maeda Y, Kinoshita T. A GPI processing phospholipase A2, PGAP6, modulates Nodal signaling in embryos by shedding CRIPTO. J Cell Biol 2016; 215:705-718. [PMID: 27881714 PMCID: PMC5147002 DOI: 10.1083/jcb.201605121] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/24/2016] [Accepted: 11/02/2016] [Indexed: 01/22/2023] Open
Abstract
Lee et al. show that PGAP6 is a glycosylphosphatidylinositol (GPI)-specific phospholipase A2 expressed on the cell surface. PGAP6 selectively acts on a GPI anchor of CRIPTO, but not its close homologue CRYPTIC, and modulates Nodal signaling during embryonic development. Glycosylphosphatidylinositol-anchored proteins (GPI-APs) can be shed from the cell membrane by GPI cleavage. In this study, we report a novel GPI-processing enzyme, termed post-glycosylphosphatidylinositol attachment to proteins 6 (PGAP6), which is a GPI-specific phospholipase A2 mainly localized at the cell surface. CRIPTO, a GPI-AP, which plays critical roles in early embryonic development by acting as a Nodal coreceptor, is a highly sensitive substrate of PGAP6, whereas CRYPTIC, a close homologue of CRIPTO, is not sensitive. CRIPTO processed by PGAP6 was released as a lysophosphatidylinositol-bearing form, which is further cleaved by phospholipase D. CRIPTO shed by PGAP6 was active as a coreceptor in Nodal signaling, whereas cell-associated CRIPTO activity was reduced when PGAP6 was expressed. Homozygous Pgap6 knockout mice showed defects in early embryonic development, particularly in the formation of the anterior–posterior axis, which are common features with Cripto knockout embryos. These results suggest PGAP6 plays a critical role in Nodal signaling modulation through CRIPTO shedding.
Collapse
Affiliation(s)
- Gun-Hee Lee
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Morihisa Fujita
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Katsuyoshi Takaoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshiko Murakami
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshitaka Fujihara
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Noriyuki Kanzawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kei-Ichi Murakami
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Eriko Kajikawa
- Center for Developmental Biology, Institute of Physical and Chemical Research, Kobe, Hyogo 650-0047, Japan
| | - Yoko Takada
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kazunobu Saito
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Hamada
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.,Center for Developmental Biology, Institute of Physical and Chemical Research, Kobe, Hyogo 650-0047, Japan
| | - Yusuke Maeda
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Taroh Kinoshita
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan .,World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
33
|
Ectopic expression of Cripto-1 in transgenic mouse embryos causes hemorrhages, fatal cardiac defects and embryonic lethality. Sci Rep 2016; 6:34501. [PMID: 27687577 PMCID: PMC5043281 DOI: 10.1038/srep34501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/15/2016] [Indexed: 12/23/2022] Open
Abstract
Targeted disruption of Cripto-1 in mice caused embryonic lethality at E7.5, whereas we unexpectedly found that ectopic Cripto-1 expression in mouse embryos also led to embryonic lethality, which prompted us to characterize the causes and mechanisms underlying embryonic death due to ectopic Cripto-1 expression. RCLG/EIIa-Cre embryos displayed complex phenotypes between embryonic day 14.5 (E14.5) and E17.5, including fatal hemorrhages (E14.5-E15.5), embryo resorption (E14.5-E17.5), pale body surface (E14.5-E16.5) and no abnormal appearance (E14.5-E16.5). Macroscopic and histological examination revealed that ectopic expression of Cripto-1 transgene in RCLG/EIIa-Cre embryos resulted in lethal cardiac defects, as evidenced by cardiac malformations, myocardial thinning, failed assembly of striated myofibrils and lack of heartbeat. In addition, Cripto-1 transgene activation beginning after E8.5 also caused the aforementioned lethal cardiac defects in mouse embryos. Furthermore, ectopic Cripto-1 expression in embryonic hearts reduced the expression of cardiac transcription factors, which is at least partially responsible for the aforementioned lethal cardiac defects. Our results suggest that hemorrhages and cardiac abnormalities are two important lethal factors in Cripto-1 transgenic mice. Taken together, these findings are the first to demonstrate that sustained Cripto-1 transgene expression after E11.5 causes fatal hemorrhages and lethal cardiac defects, leading to embryonic death at E14.5-17.5.
Collapse
|
34
|
Zhao Y, Yan H, Qiao S, Zhang L, Wang T, Meng Q, Chen X, Lin FH, Guo K, Li C, Tian W. Hydrogels bearing bioengineered mimetic embryonic microenvironments for tumor reversion. J Mater Chem B 2016; 4:6183-6191. [PMID: 32263630 DOI: 10.1039/c6tb00927a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Embryonic microenvironments can reverse the metastatic phenotype of aggressive tumors by inhibiting the Nodal signaling pathway. Here, we hypothesize that embryonic microenvironments can be transplanted for the purpose of oncotherapy. We report the development of an injectable bioactive hydrogel system containing the key antagonists of Nodal signaling-Cripto-1 receptor antibodies (2B11)-for the creation of embryonic microenvironments and the examination of their effect on tumor reversion treatment using a mouse model. Our in vitro results show that the hydrogel system can reduce the mitochondrial membrane potential of MDA-MB-231 and MCF-7, promote cell apoptosis, and reduce the invasive ability of cells. Our in vivo results illustrate that the hydrogel system can significantly inhibit tumor growth in both breast cancer and melanoma tumor-bearing mouse models, as well as transform the cell morphology of melanoma B16 cells to melanin-like cells. Furthermore, the results of the up-regulation of tumor suppressor genes and the down-regulation of oncogenes by high-throughput sequencing confirm that the developed system can also selectively turn on some tumor suppressor genes and turn off certain oncogenes so as to prompt the benign reversion of the tumor phenotype. Taken together, our results demonstrate the injectable biomaterial system is able to create an effective microenvironment for melanoma and breast tumor therapy.
Collapse
Affiliation(s)
- Yufang Zhao
- Bio-X Center, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, P. R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Castro NP, Fedorova-Abrams ND, Merchant AS, Rangel MC, Nagaoka T, Karasawa H, Klauzinska M, Hewitt SM, Biswas K, Sharan SK, Salomon DS. Cripto-1 as a novel therapeutic target for triple negative breast cancer. Oncotarget 2016; 6:11910-29. [PMID: 26059540 PMCID: PMC4494913 DOI: 10.18632/oncotarget.4182] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/09/2015] [Indexed: 12/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) presents the poorest prognosis among the breast cancer subtypes and no current standard therapy. Here, we performed an in-depth molecular analysis of a mouse model that establishes spontaneous lung metastasis from JygMC(A) cells. These primary tumors resembled the triple-negative breast cancer (TNBC) both phenotypically and molecularly. Morphologically, primary tumors presented both epithelial and spindle-like cells but displayed only adenocarcinoma-like features in lung parenchyma. The use of laser-capture microdissection combined with Nanostring mRNA and microRNA analysis revealed overexpression of either epithelial and miRNA-200 family or mesenchymal markers in adenocarcinoma and mesenchymal regions, respectively. Cripto-1, an embryonic stem cell marker, was present in spindle-like areas and its promoter showed activity in primary tumors. Cripto-1 knockout by the CRISPR-Cas9 system inhibited tumor growth and pulmonary metastasis. Our findings show characterization of a novel mouse model that mimics the TNBC and reveal Cripto-1 as a TNBC target hence may offer alternative treatment strategies for TNBC.
Collapse
Affiliation(s)
- Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | | | - Anand S Merchant
- CCRIFX Bioinformatics Core, National Cancer Institute, Bethesda, MD, USA
| | - Maria Cristina Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Tadahiro Nagaoka
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Hideaki Karasawa
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Malgorzata Klauzinska
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kajal Biswas
- Genetics of Cancer Susceptibility Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Shyam K Sharan
- Genetics of Cancer Susceptibility Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
36
|
Rangel MC, Bertolette D, Castro NP, Klauzinska M, Cuttitta F, Salomon DS. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Res Treat 2016; 156:211-26. [PMID: 26968398 PMCID: PMC4819564 DOI: 10.1007/s10549-016-3746-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Cancer has been considered as temporal and spatial aberrations of normal development in tissues. Similarities between mammary embryonic development and cell transformation suggest that the underlying processes required for mammary gland development are also those perturbed during various stages of mammary tumorigenesis and breast cancer (BC) development. The master regulators of embryonic development Cripto-1, Notch/CSL, and Wnt/β-catenin play key roles in modulating mammary gland morphogenesis and cell fate specification in the embryo through fetal mammary stem cells (fMaSC) and in the adult organism particularly within the adult mammary stem cells (aMaSC), which determine mammary progenitor cell lineages that generate the basal/myoepithelial and luminal compartments of the adult mammary gland. Together with recognized transcription factors and embryonic stem cell markers, these embryonic regulatory molecules can be inappropriately augmented during tumorigenesis to support the tumor-initiating cell (TIC)/cancer stem cell (CSC) compartment, and the effects of their deregulation may contribute for the etiology of BC, in particular the most aggressive subtype of BC, triple-negative breast cancer (TNBC). This in depth review will present evidence of the involvement of Cripto-1, Notch/CSL, and Wnt/β-catenin in the normal mammary gland morphogenesis and tumorigenesis, from fMaSC/aMaSC regulation to TIC generation and maintenance in TNBC. Specific therapies for treating TNBC by targeting these embryonic pathways in TICs will be further discussed, providing new opportunities to destroy not only the bulk tumor, but also TICs that initiate and promote the metastatic spread and recurrence of this aggressive subtype of BC.
Collapse
Affiliation(s)
- Maria Cristina Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Daniel Bertolette
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Malgorzata Klauzinska
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - Frank Cuttitta
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Building 560, Room 32-40B, 1050 Boyles Street, Ft. Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
37
|
Serum cripto-1 as a clinical marker for lung cancer. Int J Biol Markers 2015; 30:e369-73. [PMID: 26109366 DOI: 10.5301/jbm.5000155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND Cripto-1 (CR-1) plays an important role in angiogenesis related to tumor growth, in concert with vascular endothelial growth factor (VEGF), and enhanced expression of CR-1 has been reported in lung cancer tissue. METHODS Patients with lung cancer (n = 156) and healthy volunteers (n = 60) were enrolled in the study. Serum CR-1 and VEGF concentrations were measured using enzyme-linked immunosorbent assay. RESULTS Patients with lung cancer had higher serum CR-1 (4.03 ± 1.49 ng/mL vs. 1.13 ± 0.43 ng/mL, p<0.05) and VEGF (503.62 ± 112.74 pg/mL vs. 398.62 ± 117.84 pg/mL, p<0.05) levels than the control group. With stage progression in non-small cell lung cancer (NSCLC), serum CR-1 levels increased, and patients with distant metastasis had higher levels than those without metastasis (p<0.05). In NSCLC, the low CR-1 group (<1.54 ng/mL) had a better overall survival rate compared with the high CR-1 group (>1.54 ng/mL, p<0.05). CONCLUSIONS Our study suggests that serum CR-1 is a useful diagnostic and prognostic marker for NSCLC patients.
Collapse
|
38
|
Cripto-1 Promotes the Epithelial-Mesenchymal Transition in Esophageal Squamous Cell Carcinoma Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:421285. [PMID: 26472984 PMCID: PMC4589627 DOI: 10.1155/2015/421285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/14/2015] [Indexed: 12/02/2022]
Abstract
Esophageal carcinoma is a major public health problem worldwide and one of the most aggressively malignant neoplasms. Although considerable diagnostic and therapeutic progress has been made in recent years, the prognosis of EC patients still remains dismal due to high rates of recurrence/metastasis and invasion. Previous studies have demonstrated that Epithelial mesenchymal transition (EMT) is proposed as a critical mechanism for the acquisition of malignant phenotypes by epithelial cells. Several lines of evidence have shown that Cripto-1 plays an important oncogenic role during tumorigenesis by promoting EMT. The aim of our study was to evaluate the significance of Cripto-1 which plays a role in EMT and its metastasis in esophageal carcinoma. Data of this study suggest that Cripto-1 overexpression is connected with the tumorigenesis and progression of esophageal carcinoma; shRNA might be feasible for the inhibition of the invasion and metastasis of esophageal carcinoma.
Collapse
|
39
|
Cripto-1 expression and its prognostic value in human bladder cancer patients. Tumour Biol 2014; 36:1105-13. [DOI: 10.1007/s13277-014-2695-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/30/2014] [Indexed: 11/25/2022] Open
|
40
|
The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition. Semin Cancer Biol 2014; 29:51-8. [PMID: 25153355 DOI: 10.1016/j.semcancer.2014.08.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/07/2014] [Indexed: 01/04/2023]
Abstract
Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.
Collapse
|
41
|
Giorgio E, Liguoro A, D'Orsi L, Mancinelli S, Barbieri A, Palma G, Arra C, Liguori GL. Cripto haploinsufficiency affects in vivo colon tumor development. Int J Oncol 2014; 45:31-40. [PMID: 24805056 PMCID: PMC4079161 DOI: 10.3892/ijo.2014.2412] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/12/2014] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common and aggressive cancers arising from alterations in various signaling pathways, such as the WNT, RAS-MAPK, PI3K and transforming growth factor-β (TGF-β) pathways. Cripto (also called Teratocarcinoma-derived growth factor), the original member of the vertebrate EGF-CFC family, plays a key role in all of these pathways and is deeply involved in early embryo development and cancer progression. The role of Cripto in colon and breast cancer, in particular, has been investigated, as it is still not clearly understood. In this article, we provide the first in vivo functional evidence of a role of Cripto in colon cancer development. We analyzed the effect of Cripto haploinsufficiency on colon tumor formation by treating Cripto heterozygous mice with the colonotropic carcinogen azoxymethane (AOM). Of note, in our model system, Cripto haploinsufficiency increased tumorigenesis. Moreover, we revealed a correlation between the differential AOM response found in wt and Cripto⁺/⁻ mice and the expression levels of glucose regulated protein-78 (Grp78), a heat shock protein required for Cripto signaling pathways. We hypothesize that the balance between Cripto and Grp78 expression levels might be crucial in cancer development and may account for the increased tumorigenesis in Cripto heterozygous mice. In summary, our results highlight the heterogeneous effect of Cripto on tumorigenesis and the consequent high level of complexity in the Cripto regulatory pathway, whose imbalance causes tumors.
Collapse
Affiliation(s)
- Emilia Giorgio
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Annamaria Liguoro
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Luca D'Orsi
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Sara Mancinelli
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Antonio Barbieri
- Istituto Nazionale per lo studio e la cura dei Tumori IRCCS 'Fondazione G. Pascale', 80131 Naples, Italy
| | - Giuseppe Palma
- Istituto Nazionale per lo studio e la cura dei Tumori IRCCS 'Fondazione G. Pascale', 80131 Naples, Italy
| | - Claudio Arra
- Istituto Nazionale per lo studio e la cura dei Tumori IRCCS 'Fondazione G. Pascale', 80131 Naples, Italy
| | - Giovanna L Liguori
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| |
Collapse
|
42
|
Signal peptidase complex 18, encoded by SEC11A, contributes to progression via TGF-α secretion in gastric cancer. Oncogene 2013; 33:3918-26. [PMID: 23995782 DOI: 10.1038/onc.2013.364] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/04/2013] [Accepted: 07/16/2013] [Indexed: 12/26/2022]
Abstract
We built an in-house oligonucleotide array on which 394 genes were selected based on our Serial Analysis of Gene Expression (SAGE) data and previously reported array data and listed several genes related to cancer progression. Among these, we focused on SEC11A, which encodes the SPC18 protein. SEC11A mRNA expression was measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) in gastric cancer (GC) tissue samples. Expression and distribution of SPC18 protein were investigated by immunohistochemical analysis in two independent GC cohorts (Hiroshima cohort, n=99 and Chiba cohort, n=989). To determine the effect of SPC18 on cell viability and invasiveness in vitro, MTT and Boyden chamber invasion assays were performed. To evaluate the influence of SPC18 on cell growth in vivo, GC cells were injected into severe combined immunodeficiency mice. Levels of TGF-α and EGF in media from the GC cells were measured by enzyme-linked immunosorbent assay (ELISA). Studies in human tissue revealed overexpression of SEC11A mRNA in 40% of 42 GC samples by qRT-PCR. Immunohistochemical analysis of SPC18 revealed that 26 and 20% of GC cases were SPC18-positive in the Hiroshima and Chiba cohorts, respectively. In both cohorts, the Kaplan-Meier analysis showed poorer survival in SPC18-positive GC cases than in SPC18-negative GC cases. Forced expression of SPC18 activates GC cell growth in vitro and in vivo. The levels of TGF-α in culture media from GC cells were reduced by knockdown of SPC18. These results indicate that SPC18 contributes to malignant progression through promotion of TGF-α secretion in GC.
Collapse
|
43
|
MicroRNA miR-98 inhibits tumor angiogenesis and invasion by targeting activin receptor-like kinase-4 and matrix metalloproteinase-11. Oncotarget 2013; 3:1370-85. [PMID: 23211491 PMCID: PMC3717799 DOI: 10.18632/oncotarget.717] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis and invasion are essential processes for solid tumor growth and dissemination. The tumor development process can be dependent on the activation of a series of signaling pathways, including growth factor-activated pathways. MicroRNAs have been shown to be critical for tumorigenesis, but their roles in cancer angiogenesis, invasion and other signaling pathways important for tumor development are still unclear in the context of tumor biology. We investigated the role of microRNA miR-98 in regulating tumor growth, invasion, and angiogenesis using a highly aggressive breast cancer model in vitro and in vitro. We found that the expression of miR-98 inhibited breast cancer cell proliferation, survival, tumor growth, invasion, and angiogenesis. Conversely, inhibition of endogenous miR-98 promoted cell proliferation, survival, tumor growth, invasion, and angiogenesis. It appeared that miR-98 inhibited angiogenesis by modulating endothelial cell activities including cell spreading, cell invasion and tubule formation. Interestingly, miR-98 reduced the expression of ALK4 and MMP11, both of which were potential targets of miR-98. Transfection of an anti-miR-98 construct increased the expression of both targets. We confirmed that mir-98 targeted the 3'-untranslated regions of ALK4 and MMP11. Finally, ALK4- and MMP11-specific siRNAs inhibited breast cancer cell proliferation, survival, and angiogenesis. Rescue experiments with ALK4 and MMP11 constructs reversed the anti-proliferative, anti-invasive and anti-angiogenic effects of miR-98. Our findings define a regulatory role of miR-98 in tumor angiogenesis and invasion through repressed ALK4 and MMP11 expression.
Collapse
|
44
|
Liu Y, Mukhopadhyay P, Pisano MM, Lu X, Huang L, Lu Q, Dean DC. Repression of Zeb1 and hypoxia cause sequential mesenchymal-to-epithelial transition and induction of aid, Oct4, and Dnmt1, leading to immortalization and multipotential reprogramming of fibroblasts in spheres. Stem Cells 2013; 31:1350-62. [PMID: 23554223 PMCID: PMC4265806 DOI: 10.1002/stem.1382] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/06/2013] [Indexed: 12/26/2022]
Abstract
In this study, we demonstrate that sphere formation triggers immortalization and stable reprogramming of mouse fibroblasts. Cell contact signaling in spheres causes downregulation of the epithelial-to-mesenchymal transition transcription factor Zeb1 leading to rapid mesenchymal-to-epithelial transition. Hypoxia within spheres together with loss of Zeb1 repression synergize to cause superinduction of Hif1a, which in turn leads to induction of the DNA demethylase Aid/Aicda, demethylation of the Oct4 promoter/enhancer and multipotency. Oct4 and Nanog expression diminish when cells are removed from the hypoxic environment of spheres and placed in monolayer culture, but the cells retain multipotential capacity, demonstrating stable reprogramming and a gene expression pattern resembling adult stem cells. Oct4 has been shown to induce Dnmt1 in mesenchymal stem cells, and we link Oct4 and Dnmt1 to silencing of cell cycle inhibitory cyclin dependent kinase inhibitors and Arf, and immortalization of the reprogrammed fibroblasts. Sphere formation then represents a novel and rapid protocol for immortalization and stable reprogramming of fibroblasts to multipotency that does not require exogenous expression of a stem cell factor or a lineage-specifying transcription factor.
Collapse
Affiliation(s)
- Yongqing Liu
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Partha Mukhopadhyay
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - M. Michele Pisano
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Xiaoqin Lu
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Li Huang
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- School of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Qingxian Lu
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Douglas C. Dean
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Ophthalmology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Department of Biochemistry and Molecular Biology, University of Louisville Health Sciences Center, Louisville, KY 40202
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202
| |
Collapse
|
45
|
Bianco C, Castro NP, Baraty C, Rollman K, Held N, Rangel MC, Karasawa H, Gonzales M, Strizzi L, Salomon DS. Regulation of human Cripto-1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells. J Cell Physiol 2013; 228:1174-88. [PMID: 23129342 PMCID: PMC3573215 DOI: 10.1002/jcp.24271] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 10/18/2012] [Indexed: 11/07/2022]
Abstract
Human Cripto-1 (CR-1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR-1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)-1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR-1 gene expression in NTERA-2 human embryonal carcinoma cells and in breast cancer cells. CR-1 expression in NTERA-2 cells was positively regulated by LRH-1 through direct binding to a DR0 element within the CR-1 promoter, while GCNF strongly suppressed CR-1 expression in these cells. In addition, the CR-1 promoter was unmethylated in NTERA-2 cells, while T47D, ZR75-1, and MCF7 breast cancer cells showed high levels of CR-1 promoter methylation and low CR-1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR-1 promoter and reactivated CR-1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR-1 was highly expressed in the majority of human breast tumors, suggesting that CR-1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR-1 gene expression and its critical role in the pathogenesis of human cancer.
Collapse
MESH Headings
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Binding Sites
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Embryonal/genetics
- Carcinoma, Embryonal/metabolism
- Carcinoma, Embryonal/pathology
- Cell Movement
- DNA Methylation/drug effects
- DNA Modification Methylases/antagonists & inhibitors
- DNA Modification Methylases/metabolism
- Decitabine
- Dose-Response Relationship, Drug
- Embryonal Carcinoma Stem Cells/metabolism
- Embryonal Carcinoma Stem Cells/pathology
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Histone Deacetylase Inhibitors/pharmacology
- Humans
- Hydroxamic Acids/pharmacology
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Luciferases/biosynthesis
- Luciferases/genetics
- MCF-7 Cells
- Neoplasm Invasiveness
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Receptor Subfamily 6, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism
- Promoter Regions, Genetic
- RNA Interference
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Time Factors
- Tissue Array Analysis
- Transcription, Genetic
- Transfection
- Tretinoin/pharmacology
- Valproic Acid/pharmacology
Collapse
Affiliation(s)
- Caterina Bianco
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nadia P. Castro
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Christina Baraty
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Kelly Rollman
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Natalie Held
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Maria Cristina Rangel
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Hideaki Karasawa
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Monica Gonzales
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Luigi Strizzi
- Children’s Memorial Research Center, Robert H. Lurie Comprehensive Cancer Center Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David S. Salomon
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
46
|
Nagaoka T, Karasawa H, Turbyville T, Rangel MC, Castro NP, Gonzales M, Baker A, Seno M, Lockett S, Greer YE, Rubin JS, Salomon DS, Bianco C. Cripto-1 enhances the canonical Wnt/β-catenin signaling pathway by binding to LRP5 and LRP6 co-receptors. Cell Signal 2013; 25:178-89. [PMID: 23022962 PMCID: PMC3508164 DOI: 10.1016/j.cellsig.2012.09.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/18/2012] [Accepted: 09/24/2012] [Indexed: 12/21/2022]
Abstract
Cripto-1 is implicated in multiple cellular events, including cell proliferation, motility and angiogenesis, through the activation of an intricate network of signaling pathways. A crosstalk between Cripto-1 and the canonical Wnt/β-catenin signaling pathway has been previously described. In fact, Cripto-1 is a downstream target gene of the canonical Wnt/β-catenin signaling pathway in the embryo and in colon cancer cells and T-cell factor (Tcf)/lymphoid enhancer factor binding sites have been identified in the promoter and the first intronic region of the mouse and human Cripto-1 genes. We now demonstrate that Cripto-1 modulates signaling through the canonical Wnt/β-catenin/Tcf pathway by binding to the Wnt co-receptors low-density lipoprotein receptor-related protein (LRP) 5 and LRP6, which facilitates Wnt3a binding to LRP5 and LRP6. Cripto-1 functionally enhances Wnt3a signaling through cytoplasmic stabilization of β-catenin and elevated β-catenin/Tcf transcriptional activation. Conversely, Wnt3a further increases Cripto-1 stimulation of migration, invasion and colony formation in soft agar of HC11 mouse mammary epithelial cells, indicating that Cripto-1 and the canonical Wnt/β-catenin signaling co-operate in regulating motility and in vitro transformation of mammary epithelial cells.
Collapse
Affiliation(s)
- Tadahiro Nagaoka
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Hideaki Karasawa
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Thomas Turbyville
- Optical Microscopy and Analysis Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Maria-Cristina Rangel
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Nadia P. Castro
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Monica Gonzales
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Alyson Baker
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Masaharu Seno
- Laboratory of Nano-Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushima-Naka, Okayama 700-8530, Japan
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, USA
| | - Yoshimi E. Greer
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Bldg 37/Room 2066, Bethesda, MD 20892, USA
| | - Jeffrey S. Rubin
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Bldg 37/Room 2066, Bethesda, MD 20892, USA
| | - David S. Salomon
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| | - Caterina Bianco
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, 1050 Boyles St., Bldg 560/ Room 12-46, Frederick, MD 21702, USA
| |
Collapse
|
47
|
Rangel MC, Karasawa H, Castro NP, Nagaoka T, Salomon DS, Bianco C. Role of Cripto-1 during epithelial-to-mesenchymal transition in development and cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2188-200. [PMID: 22542493 DOI: 10.1016/j.ajpath.2012.02.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 02/13/2012] [Accepted: 02/21/2012] [Indexed: 02/08/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a critical multistep process that converts epithelial cells to more motile and invasive mesenchymal cells, contributing to body patterning and morphogenesis during embryonic development. In addition, both epithelial plasticity and increased motility and invasiveness are essential for the branching morphogenesis that occurs during development of the mammary gland and during tumor formation, allowing cancer cells to escape from the primary tumor. Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic (EGF/CFC) gene family, together with the transforming growth factor (TGF)-β family ligand Nodal, regulates both cell movement and EMT during embryonic development. During postnatal development, Cripto-1 regulates the branching morphogenesis of the mouse mammary gland and enhances both the invasive and migratory properties of mammary epithelial cells in vitro. Furthermore, transgenic mouse models have shown that Cripto-1 promotes the formation of mammary tumors that display properties of EMT, including the down-regulation of the cell surface adherens junctional protein E-cadherin and the up-regulation of mesenchymal markers, such as vimentin, N-cadherin, and Snail. Interestingly, Cripto-1 is enriched in a subpopulation of embryonal, melanoma, prostate, and pancreatic cancer cells that possess stem-like characteristics. Therefore, Cripto-1 may play a role during developmental EMT, and it may also be involved in the reprogramming of differentiated tumor cells into cancer stem cells through the induction of an EMT program.
Collapse
Affiliation(s)
- Maria C Rangel
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
48
|
Nagaoka T, Karasawa H, Castro NP, Rangel MC, Salomon DS, Bianco C. An evolving web of signaling networks regulated by Cripto-1. Growth Factors 2012; 30:13-21. [PMID: 22149969 DOI: 10.3109/08977194.2011.641962] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Over the past few decades, our understanding of the embryonic gene Cripto-1 has considerably advanced through biochemical, cell biology, and animal studies. Cripto-1 performs key functions during embryonic development, while it dramatically disappears in adult tissues, except possibly in adult tissue stem cells. Cripto-1 is re-expressed in human tumors promoting cell proliferation, migration, invasion, epithelial to mesenchymal transition, and tumor angiogenesis. This diversity of biological effects is dependent upon interaction of Cripto-1 with an extensive array of signaling molecules. In fact, Cripto-1 modulates signaling of transforming growth factor-β family members, including Nodal, GDF-1/-3, Activin, and TGF-β1, activates c-src/MAPK/Protein Kinase B (AKT) pathway in a Glypican-1 and GRP78-dependent manner, and cross-talks with erbB4, Wnt/β-catenin, Notch, Caveolin-1, and Apelin/putative receptor protein related to Angiotensin-type I receptor (APJ) pathways. This article provides an updated survey of the various signaling pathways modulated by Cripto-1 with a focus on mechanistic insights in our understanding of the biological function of Cripto-1 in eukaryotic cells.
Collapse
Affiliation(s)
- Tadahiro Nagaoka
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
49
|
Das AB, Loying P, Bose B. Human recombinant Cripto-1 increases doubling time and reduces proliferation of HeLa cells independent of pro-proliferation pathways. Cancer Lett 2011; 318:189-98. [PMID: 22182448 DOI: 10.1016/j.canlet.2011.12.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 10/26/2011] [Accepted: 12/07/2011] [Indexed: 12/26/2022]
Abstract
Human oncofetal protein Cripto-1 (CR-1) is overexpressed in many types of cancers. CR-1 binds to cell surface Glypican-1 to activate Erk1/2 MAPK and Akt pathways leading to cell proliferation. However, we show that treatment with recombinant CR-1 reduces proliferation of HeLa cells by increasing the doubling time without triggering cell death or cell cycle arrest. Using a comparative study with U-87 MG cells, we show that the pro-proliferative pathway of CR-1 is not effective in HeLa cells due to lower expression of Glypican-1. Further we show that treatment with recombinant CR-1 increases PTEN in HeLa cells leading to downregulation of PI3K/Akt pathway. The anti-proliferative effect gets potentiated when the pro-proliferative pathway is blocked.
Collapse
Affiliation(s)
- Asim Bikas Das
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, India.
| | | | | |
Collapse
|
50
|
Galat A. Common structural traits for cystine knot domain of the TGFβ superfamily of proteins and three-fingered ectodomain of their cellular receptors. Cell Mol Life Sci 2011; 68:3437-51. [PMID: 21369710 PMCID: PMC11114550 DOI: 10.1007/s00018-011-0643-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 01/08/2011] [Accepted: 02/15/2011] [Indexed: 01/22/2023]
Abstract
The transforming growth factor-β (TGFβ) superfamily of proteins and their receptors are crucial developmental factors for all metazoan organisms. Cystine-knot (CK) motif is a spatial feature of the TGFβ superfamily of proteins whereas the extra-cellular domains (ectodomains) of their respective receptors form three-fingered protein domain (TFPD), both stabilized by tight cystine networks. Analyses of multiple sequence alignments of these two domains encoded in various genomes revealed that the cystines forming the CK and TFPD folds are conserved, whereas the remaining polypeptide patches are diversified. Orthologues of the human TGFβs and their respective receptors expressed in diverse vertebrates retain high sequence conservation. Examination of 3D structures of various TGFβ factors bound to their receptors have revealed that the CK and TFPD domains display several similar spatial traits suggesting that these two different protein folds might have been acquired from a common ancestor.
Collapse
Affiliation(s)
- A Galat
- Commissariat à l'Energie Atomique, Direction des Sciences du Vivant, Institut de Biologie et de Technologies de Saclay, Service d'Ingénierie Moléculaire des Protéines, Bat. 152, 91191 Gif-sur-Yvette, France.
| |
Collapse
|