1
|
Nakanoh S, Sham K, Ghimire S, Mohorianu I, Rayon T, Vallier L. Human surface ectoderm and amniotic ectoderm are sequentially specified according to cellular density. SCIENCE ADVANCES 2024; 10:eadh7748. [PMID: 38427729 PMCID: PMC10906920 DOI: 10.1126/sciadv.adh7748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Mechanisms specifying amniotic ectoderm and surface ectoderm are unresolved in humans due to their close similarities in expression patterns and signal requirements. This lack of knowledge hinders the development of protocols to accurately model human embryogenesis. Here, we developed a human pluripotent stem cell model to investigate the divergence between amniotic and surface ectoderms. In the established culture system, cells differentiated into functional amnioblast-like cells. Single-cell RNA sequencing analyses of amnioblast differentiation revealed an intermediate cell state with enhanced surface ectoderm gene expression. Furthermore, when the differentiation started at the confluent condition, cells retained the expression profile of surface ectoderm. Collectively, we propose that human amniotic ectoderm and surface ectoderm are specified along a common nonneural ectoderm trajectory based on cell density. Our culture system also generated extraembryonic mesoderm-like cells from the primed pluripotent state. Together, this study provides an integrative understanding of the human nonneural ectoderm development and a model for embryonic and extraembryonic human development around gastrulation.
Collapse
Affiliation(s)
- Shota Nakanoh
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge CB22 3AT, UK
| | - Kendig Sham
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sabitri Ghimire
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Irina Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Teresa Rayon
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge CB22 3AT, UK
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Berlin Institute of Health Centre for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
- Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| |
Collapse
|
2
|
Ucci S, Spaziani S, Quero G, Vaiano P, Principe M, Micco A, Sandomenico A, Ruvo M, Consales M, Cusano A. Advanced Lab-on-Fiber Optrodes Assisted by Oriented Antibody Immobilization Strategy. BIOSENSORS 2022; 12:1040. [PMID: 36421158 PMCID: PMC9688615 DOI: 10.3390/bios12111040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Lab-on-fiber (LoF) optrodes offer several advantages over conventional techniques for point-of-care platforms aimed at real-time and label-free detection of clinically relevant biomarkers. Moreover, the easy integration of LoF platforms in medical needles, catheters, and nano endoscopes offer unique potentials for in vivo biopsies and tumor microenvironment assessment. The main barrier to translating the vision close to reality is the need to further lower the final limit of detection of developed optrodes. For immune-biosensing purposes, the assay sensitivity significantly relies on the capability to correctly immobilize the capture antibody in terms of uniform coverage and correct orientation of the bioreceptor, especially when very low detection limits are requested as in the case of cancer diagnostics. Here, we investigated the possibility to improve the immobilization strategies through the use of hinge carbohydrates by involving homemade antibodies that demonstrated a significantly improved recognition of the antigen with ultra-low detection limits. In order to create an effective pipeline for the improvement of biofunctionalization protocols to be used in connection with LoF platforms, we first optimized the protocol using a microfluidic surface plasmon resonance (mSPR) device and then transferred the optimized strategy onto LoF platforms selected for the final validation. Here, we selected two different LoF platforms: a biolayer interferometry (BLI)-based device (commercially available) and a homemade advanced LoF biosensor based on optical fiber meta-tips (OFMTs). As a clinically relevant scenario, here we focused our attention on a promising serological biomarker, Cripto-1, for its ability to promote tumorigenesis in breast and liver cancer. Currently, Cripto-1 detection relies on laborious and time-consuming immunoassays. The reported results demonstrated that the proposed approach based on oriented antibody immobilization was able to significantly improve Cripto-1 detection with a 10-fold enhancement versus the random approach. More interestingly, by using the oriented antibody immobilization strategy, the OFMTs-based platform was able to reveal Cripto-1 at a concentration of 0.05 nM, exhibiting detection capabilities much higher (by a factor of 250) than those provided by the commercial LoF platform based on BLI and similar to the ones shown by the commercial and well-established bench-top mSPR Biacore 8K system. Therefore, our work opened new avenues into the development of high-sensitivity LoF biosensors for the detection of clinically relevant biomarkers in the sub-ng/mL range.
Collapse
Affiliation(s)
- Sarassunta Ucci
- Institute of Biostructures and Bioimaging, National Research Council of Italy, Via P. Castellino, 111, 80131 Naples, Italy
| | - Sara Spaziani
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy
| | - Giuseppe Quero
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy
| | - Patrizio Vaiano
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
| | - Maria Principe
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
| | - Alberto Micco
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy
| | - Annamaria Sandomenico
- Institute of Biostructures and Bioimaging, National Research Council of Italy, Via P. Castellino, 111, 80131 Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, National Research Council of Italy, Via P. Castellino, 111, 80131 Naples, Italy
| | - Marco Consales
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy
| | - Andrea Cusano
- Optoelectronics Group, Engineering Department, University of Sannio, c.so Garibaldi 107, 82100 Benevento, Italy
- Centro Regionale Information Communication Technology (CeRICT Scrl), 82100 Benevento, Italy
| |
Collapse
|
3
|
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
|
4
|
CRIPTO-1 Is Immunolocalized in the Syncytiotrophoblast of Ampullary Pregnancies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4769790. [PMID: 35434129 PMCID: PMC9012632 DOI: 10.1155/2022/4769790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/25/2022] [Indexed: 12/02/2022]
Abstract
Introduction Controlling the invasive activity of trophoblastic tissue has not been elucidated. In the accreta placenta, the invasion of placental tissue is directly related to the expression of CRIPTO-1 at the maternal-fetal interface. The aim of this study is to evaluate if the expression of the CRIPTO-1 is related to different degrees of trophoblast invasion into the tube wall in ampullary pregnancy. Methods Prospective study with 21 patients with ampullary tubal pregnancy undergoing salpingectomy. Anatomopathological evaluation determined the degree of invasion of trophoblast tissues into the tubal wall and grouped the samples into invasive degrees I, II, or III. The groups were compared for tissue expression of CRIPTO-1 using the Kruskal-Wallis nonparametric test. p values lower than 0.05 were considered significant. Results Quantitative expression of CRIPTO-1 differed in each of the three groups of trophoblast invasion in the tubal wall in ampullary pregnancies (p < 0.001). There is a difference between groups when grade I + grade II versus grade III (p < 0.001) and grade I versus grade II + grade III (p < 0.001). The tissue expression of CRIPTO-1 in ectopic trophoblasts showed that deeper invasion of the tubal wall was associated with stronger expression than in shallow invasion (p < 0.001). Discussion. In ampullary pregnancies, the depth of penetration of trophoblast tissue in the tubal wall is related to CRIPTO-1 tissue expression.
Collapse
|
5
|
Freeman DW, Rodrigues Sousa E, Karkampouna S, Zoni E, Gray PC, Salomon DS, Kruithof-de Julio M, Spike BT. Whence CRIPTO: The Reemergence of an Oncofetal Factor in 'Wounds' That Fail to Heal. Int J Mol Sci 2021; 22:10164. [PMID: 34576327 PMCID: PMC8472190 DOI: 10.3390/ijms221810164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023] Open
Abstract
There exists a set of factors termed oncofetal proteins that play key roles in ontogeny before they decline or disappear as the organism's tissues achieve homeostasis, only to then re-emerge in cancer. Although the unique therapeutic potential presented by such factors has been recognized for more than a century, their clinical utility has yet to be fully realized1. This review highlights the small signaling protein CRIPTO encoded by the tumor derived growth factor 1 (TDGF1/Tdgf1) gene, an oft cited oncofetal protein whose presence in the cancer literature as a tumor promoter, diagnostic marker and viable therapeutic target continues to grow. We touch lightly on features well established and well-reviewed since its discovery more than 30 years ago, including CRIPTO's early developmental roles and modulation of SMAD2/3 activation by a selected set of transforming growth factor β (TGF-β) family ligands. We predominantly focus instead on more recent and less well understood additions to the CRIPTO signaling repertoire, on its potential upstream regulators and on new conceptual ground for understanding its mode of action in the multicellular and often stressful contexts of neoplastic transformation and progression. We ask whence it re-emerges in cancer and where it 'hides' between the time of its fetal activity and its oncogenic reemergence. In this regard, we examine CRIPTO's restriction to rare cells in the adult, its potential for paracrine crosstalk, and its emerging role in inflammation and tissue regeneration-roles it may reprise in tumorigenesis, acting on subsets of tumor cells to foster cancer initiation and progression. We also consider critical gaps in knowledge and resources that stand between the recent, exciting momentum in the CRIPTO field and highly actionable CRIPTO manipulation for cancer therapy and beyond.
Collapse
Affiliation(s)
- David W. Freeman
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| | - Elisa Rodrigues Sousa
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Sofia Karkampouna
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Eugenio Zoni
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
| | - Peter C. Gray
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA;
| | - David S. Salomon
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 20893, USA;
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, 3012 Bern, Switzerland; (E.R.S.); (S.K.); (E.Z.)
- Translational Organoid Models, Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, 3010 Bern, Switzerland
- Department of Urology, Inselspital, University Hospital of Bern, 3010 Bern, Switzerland
| | - Benjamin T. Spike
- Department of Oncological Sciences, School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Arboretto P, Cillo M, Leonardi A. New Insights into Cancer Targeted Therapy: Nodal and Cripto-1 as Attractive Candidates. Int J Mol Sci 2021; 22:ijms22157838. [PMID: 34360603 PMCID: PMC8345935 DOI: 10.3390/ijms22157838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor beta (TGF-β) signaling is fundamental for correct embryonic development. However, alterations of this pathway have been correlated with oncogenesis, tumor progression and sustaining of cancer stem cells (CSCs). Cripto-1 (CR-1) and Nodal are two embryonic proteins involved in TGF-β signaling. Their expression is almost undetectable in terminally differentiated cells, but they are often re-expressed in tumor cells, especially in CSCs. Moreover, cancer cells that show high levels of CR-1 and/or Nodal display more aggressive phenotypes in vitro, while in vivo their expression correlates with a worse prognosis in several human cancers. The ability to target CSCs still represents an unmet medical need for the complete eradication of certain types of tumors. Given the prognostic role and the selective expression of CR-1 and Nodal on cancer cells, they represent archetypes for targeted therapy. The aim of this review is to clarify the role of CR-1 and Nodal in cancer stem populations and to summarize the current therapeutic strategy to target CSCs using monoclonal antibodies (mAbs) or other molecular tools to interfere with these two proteins.
Collapse
|
8
|
Du T, Jiang J, Chen Y, Zhang N, Chen G, Wang X, Long X, Feng X. MiR-138-1-3p alters the stemness and radiosensitivity of tumor cells by targeting CRIPTO and the JAK2/STAT3 pathway in nasopharyngeal carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:485. [PMID: 33850882 PMCID: PMC8039661 DOI: 10.21037/atm-21-521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Tumor resistance to radiotherapy is one of the main obstacles to the clinical treatment of nasopharyngeal carcinoma (NPC). Improving the radiosensitivity of tumor cells has an important clinical significance in treatment of clinical NPC. This study aimed to identify that miR-138-1-3p as a novel therapeutic target in radioresistant NPC cells and found its targets, CRIPTO and the JAK2/STAT3 pathway. Methods Radioresistant C666-IR and HK-1R cells were derived from the NPC cell lines C666-1 and HK-1. The different microRNAs (miRNAs) and their targeting genes were analyzed between C666-1 and C666-IR cells using microarray bioinformatics. Western blot, qRT-PCR, gene transfection, Luciferase reporter assay, and confocal laser scanning microscopy were applied for the analysis of the different genes. Results MiR-138-1-3p was found to target CRIPTO, which involved in the epithelial-mesenchymal transition (EMT) and JAK2/STAT3 signaling pathways. The luciferase reporter assay confirmed that miR-138-1-3p targeted CRIPTO and downregulated the expression of CRIPTO. Furthermore, miR-138-1-3p affected the stability of the CRIPTO-GRP78 complex on the cell membrane and also reversed the radioresistant characteristics of NPC stem cells, which affected EMT and the JAK2/STAT3 signaling pathway. Conclusions The miR-138-1-3p is a small molecule that can modulate radiosensitivity in the radioresistant C666-IR and HK-1R NPC cell lines by inhibiting EMT and targeting CRIPTO to reduce the activation of the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Tao Du
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China.,Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jiahui Jiang
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Yiting Chen
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Nengwei Zhang
- Department of General Surgery, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Guanyang Chen
- Department of General Surgery, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Xingwei Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xueying Long
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Xueping Feng
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China.,Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Daraghma H, Untiveros G, Raskind A, Iaccarino E, Sandomenico A, Ruvo M, Arnouk H, Ciancio MJ, Cuevas-Nunez M, Strizzi L. The role of Nodal and Cripto-1 in human oral squamous cell carcinoma. Oral Dis 2020; 27:1137-1147. [PMID: 32916013 DOI: 10.1111/odi.13640] [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: 03/27/2020] [Revised: 08/05/2020] [Accepted: 08/31/2020] [Indexed: 12/16/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a common epithelial malignancy of the oral cavity. Nodal and Cripto-1 (CR-1) are important developmental morphogens expressed in several adult cancers and are associated with disease progression. Whether Nodal and CR-1 are simultaneously expressed in the same tumor and how this affects cancer biology are unclear. We investigate the expression and potential role of both Nodal and CR-1 in human OSCC. Immunohistochemistry results show that Nodal and CR-1 are both expressed in the same human OSCC sample and that intensity of Nodal staining is correlated with advanced-stage disease. However, this was not observed with CR-1 staining. Western blot analysis of lysates from two human OSCC line experiments shows expression of CR-1 and Nodal, and their respective signaling molecules, Src and ERK1/2. Treatment of SCC25 and SCC15 cells with both Nodal and CR-1 inhibitors simultaneously resulted in reduced cell viability and reduced levels of P-Src and P-ERK1/2. Further investigation showed that the combination treatment with both Nodal and CR-1 inhibitors was capable of reducing invasiveness of SCC25 cells. Our results show a possible role for Nodal/CR-1 function during progression of human OSCC and that targeting both proteins simultaneously may have therapeutic potential.
Collapse
Affiliation(s)
- Hussein Daraghma
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA.,Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Gustavo Untiveros
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Aleksandr Raskind
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Emanuela Iaccarino
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Annamaria Sandomenico
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Hilal Arnouk
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Mae J Ciancio
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Maria Cuevas-Nunez
- College of Dental Medicine Illinois, Midwestern University, Downers Grove, IL, USA
| | - Luigi Strizzi
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| |
Collapse
|
10
|
Chen V, Iwama E, Kim IK, Giaccone G. Serum CRIPTO does not confer drug resistance against osimertinib but is an indicator of tumor burden in non-small cell lung cancer. Lung Cancer 2020; 145:48-57. [PMID: 32408132 DOI: 10.1016/j.lungcan.2020.04.032] [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: 04/04/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND Adenocarcinoma is the most common subtype of non-small cell lung cancer (NSCLC) and often harbors oncogenic driver mutations in the epidermal growth factor receptor (EGFR). Osimertinib (AZD9291), a third generation EGFR TKI, has replaced earlier generation EGFR TKIs for first line treatment of EGFR mutant lung cancer due to its improved overall survival, longer progression free survival, and better tolerability compared to earlier generation inhibitors. However, like earlier generation EGFR TKIs, only about two thirds of patients respond, indicating an unknown mechanism of intrinsic resistance for the non-responders. We previously identified overexpression of CRIPTO as a potential mechanism of intrinsic resistance to EGFR TKIs of first and second generation. OBJECTIVE To determine if CRIPTO could promote drug resistance against the third generation EGFR-TKIs osimertinib. We also wanted to investigate whether this resistance was conferred by both membrane bound and secreted CRIPTO. Finally, we wanted to explore the potential of secreted CRIPTO as a non-invasive biomarker for EGFR-TKI resistance. MATERIALS AND METHODS HCC827 and H1975, EGFR mutant non-small cell lung carcinoma (NSCLC) cell lines, were transfected with wildtype CRIPTO, two secreted variants of CRIPTO, a membrane only version of CRIPTO, and the mock backbone vector as the control. Western blotting, immunoprecipitation, and in vitro viability experiments were performed. In vivo work was carried out in athymic nude mice; 2 × 106 CRIPTO overexpressing HCC827 cells were implanted per mouse. EGFR mutant NSCLC patient blood samples were collected before treatment with and EGFR-TKI, during response while on treatment, and at progression while on treatment. RESULTS Although both membrane bound and secreted CRIPTO forms were able to activate downstream pathways such as SRC, CRIPTO was unable to elicit resistance towards osimertinib in vitro or in vivo. CRIPTO serum levels in mice were higher in larger xenograft tumors. Furthermore, CRIPTO serum levels were higher in patients with progressing lung cancer when compared to their CRIPTO serum levels during EGFR-TKI response. CONCLUSIONS CRIPTO does not cause resistance against third generation EGFR-TKI osimertinib. CRIPTO levels in serum might be a potentially useful biomarker for tumor burden in NSCLC patients.
Collapse
Affiliation(s)
- Vincent Chen
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, United States
| | - Eiji Iwama
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, United States
| | - In-Kyu Kim
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, United States
| | - Giuseppe Giaccone
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, United States.
| |
Collapse
|
11
|
Jabbarzadeh Kaboli P, Afzalipour Khoshkbejari M, Mohammadi M, Abiri A, Mokhtarian R, Vazifemand R, Amanollahi S, Yazdi Sani S, Li M, Zhao Y, Wu X, Shen J, Cho CH, Xiao Z. Targets and mechanisms of sulforaphane derivatives obtained from cruciferous plants with special focus on breast cancer - contradictory effects and future perspectives. Biomed Pharmacother 2019; 121:109635. [PMID: 31739165 DOI: 10.1016/j.biopha.2019.109635] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common type of cancer among women. Therefore, discovery of new and effective drugs with fewer side effects is necessary to treat it. Sulforaphane (SFN) is an organosulfur compound obtained from cruciferous plants, such as broccoli and mustard, and it has the potential to treat breast cancer. Hence, it is vital to find out how SFN targets certain genes and cellular pathways in treating breast cancer. In this review, molecular targets and cellular pathways of SFN are described. Studies have shown SFN inhibits cell proliferation, causes apoptosis, stops cell cycle and has anti-oxidant activities. Increasing reactive oxygen species (ROS) produces oxidative stress, activates inflammatory transcription factors, and these result in inflammation leading to cancer. Increasing anti-oxidant potential of cells and discovering new targets to reduce ROS creation reduces oxidative stress and it eventually reduces cancer risks. In short, SFN effectively affects histone deacetylases involved in chromatin remodeling, gene expression, and Nrf2 anti-oxidant signaling. This review points to the potential of SFN to treat breast cancer as well as the importance of other new cruciferous compounds, derived from and isolated from mustard, to target Keap1 and Akt, two key regulators of cellular homeostasis.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China; Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia.
| | | | - Mahsa Mohammadi
- Department of Chemistry, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ardavan Abiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Roya Mokhtarian
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia
| | - Reza Vazifemand
- Laboratory of Virology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, 43400, Malaysia
| | - Shima Amanollahi
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia; School of Mathematical, Physical, and Natural Sciences, University of Florence, Firenze, 50134, Italy
| | - Shaghayegh Yazdi Sani
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China.
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
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: 2.7] [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
|
14
|
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: 1.0] [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
|
15
|
Focà G, Iaccarino E, Focà A, Sanguigno L, Untiveros G, Cuevas-Nunez M, Strizzi L, Leonardi A, Ruvo M, Sandomenico A. Development of conformational antibodies targeting Cripto-1 with neutralizing effects in vitro. Biochimie 2019; 158:246-256. [PMID: 30703478 DOI: 10.1016/j.biochi.2019.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/22/2019] [Indexed: 01/14/2023]
Abstract
Human Cripto-1 (Cripto-1), the founding member of the EGF-CFC superfamily, is a key regulator of many processes during embryonic development and oncogenesis. Cripto-1 is barely present or even absent in normal adult tissues while it is aberrantly re-expressed in various tumors. Blockade of the CFC domain-mediated Cripto-1 functions is acknowledged as a promising therapeutic intervention point to inhibit the tumorigenic activity of the protein. In this work, we report the generation and characterization of murine monoclonal antibodies raised against the synthetic folded CFC [112-150] domain of the human protein. Through subtractive ELISA assays clones were screened for the ability to specifically recognize "hot spot" residues on the CFC domain, which are crucial for the interaction with Activin Type I receptor (ALK4) and GRP78. On selected antibodies, SPR and epitope mapping studies have confirmed their specificity and have revealed that recognition occurs only on a conformational epitope. Furthermore, FACS analyses have confirmed the ability of 1B4 antibody to recognize the membrane-anchored and soluble native Cripto-1 protein in a panel of human cancer cells. Finally, we have evaluated its functional effects through in vitro cellular signaling assays and cell cycle analysis. These findings suggest that the selected anti-CFC mAbs have the potential to neutralize the protein oncogenic activity and may be used as theranostic molecules suitable as tumor homing agents for Cripto-1-overexpressing cancer cells and tissues and to overcome drug-resistance in routine cancer therapies.
Collapse
Affiliation(s)
- Giuseppina Focà
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Emanuela Iaccarino
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Annalia Focà
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Luca Sanguigno
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Gustavo Untiveros
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA
| | - Maria Cuevas-Nunez
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA; College of Dental Medicine, Dwners Grove, Chicago, IL, USA
| | - Luigi Strizzi
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA
| | - Antonio Leonardi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy.
| | - Annamaria Sandomenico
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy.
| |
Collapse
|
16
|
Chikaraishi K, Takenobu H, Sugino RP, Mukae K, Akter J, Haruta M, Kurosumi M, Endo TA, Koseki H, Shimojo N, Ohira M, Kamijo T. CFC1 is a cancer stemness-regulating factor in neuroblastoma. Oncotarget 2018; 8:45046-45059. [PMID: 28620148 PMCID: PMC5542166 DOI: 10.18632/oncotarget.18464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/28/2017] [Indexed: 01/06/2023] Open
Abstract
Background Despite the use of aggressive therapy, survival rates among high-risk neuroblastoma (NB) patients remain poor. Cancer stem cells (CSCs) are considered to be critically involved in the recurrence and metastasis of NB and are isolated as NB spheres. Methods The gene expression profiling of adherent (control) and sphere-forming primary NB cells was conducted using a gene expression microarray. CFC1, which functions in the development of embryos and decides the left-right axis, was strongly expressed in sphere-forming cells only and was related to the unfavorable prognosis of NB patients. The knockdown and overexpression of CFC1 were performed using a lentiviral system in NB cell lines. Sphere formation, cell proliferation, colony formation in soft agar, and xenograft tumor formation were analyzed. Results The overexpression of CFC1 increased sphere formation, cell growth, and colony formation. These phenotypes, particularly sphere formation, and xenograft tumor formation were significantly suppressed by the knockdown of CFC1. CFC1 inhibited Activin A-induced NB cell differentiation and Smad2 phosphorylation in NB cell lines, indicating its involvement in tumorigenesis related to EGF-CFC co-receptor family molecule pathways. Collectively, these results indicate that CFC1 is a candidate molecule for the development of CSC-targeted therapy for NB.
Collapse
Affiliation(s)
- Koji Chikaraishi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Department of Pediatrics, Chiba University, Chiba, Japan
| | - Hisanori Takenobu
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Ryuichi P Sugino
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Kyosuke Mukae
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Jesmin Akter
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Masayuki Haruta
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | | | - Takaho A Endo
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Naoki Shimojo
- Department of Pediatrics, Chiba University, Chiba, Japan
| | - Miki Ohira
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| |
Collapse
|
17
|
Calvanese L, Focà A, Sandomenico A, Focà G, Caporale A, Doti N, Iaccarino E, Leonardi A, D'Auria G, Ruvo M, Falcigno L. Structural insights into the interaction of a monoclonal antibody and Nodal peptides by STD-NMR spectroscopy. Bioorg Med Chem 2017; 25:6589-6596. [PMID: 29113739 DOI: 10.1016/j.bmc.2017.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/05/2017] [Accepted: 10/26/2017] [Indexed: 12/31/2022]
Abstract
Nodal is a growth factor expressed during early embryonic development, but reactivated in several advanced-stage cancers. Targeting of Nodal signaling, which occurs via the binding to Cripto-1 co-receptor, results in inhibition of cell aggressiveness and reduced tumor growth. The Nodal binding region to Cripto-1 was identified and targeted with a high affinity monoclonal antibody (3D1). By STD-NMR technique, we investigated the interaction of Nodal fragments with 3D1 with the aim to elucidate at atomic level the interaction surface. Data indicate with high accuracy the antibody-antigen contact atoms and confirm the information previously obtained by immune-enzymatic methods. Main residues contacted by 3D1 are P46, V47, E49 and E50, which belong to the Nodal loop involved in the interaction with the co-receptor.
Collapse
Affiliation(s)
- Luisa Calvanese
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annalia Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Annamaria Sandomenico
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Giuseppina Focà
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Andrea Caporale
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Emanuela Iaccarino
- DISTABIF, Università degli Studi della Campania "Lugi Vanvitelli", via Vivaldi, 43, 80100 Caserta, Italy
| | - Antonio Leonardi
- Dept. Medicina Molecolare e Biotecnologie Mediche, Università Federico II di Napoli, Naples, Italy
| | - Gabriella D'Auria
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy.
| | - Lucia Falcigno
- CIRPeB, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy; Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, via Mezzocannone, 16, 80134 Napoli, Italy; Dept. of Pharmacy, University of Naples Federico II, via Mezzocannone, 16, 80134 Napoli, Italy.
| |
Collapse
|
18
|
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: 32] [Impact Index Per Article: 4.0] [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
|
19
|
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: 2.4] [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
|
20
|
Clinical significance of cripto-1 expression in lung adenocarcinoma. Oncotarget 2017; 8:79087-79098. [PMID: 29108289 PMCID: PMC5668022 DOI: 10.18632/oncotarget.15761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 02/20/2017] [Indexed: 01/15/2023] Open
Abstract
Cripto-1 can promote tumourigenesis and may be a potential prognostic biomarker in several malignancies, yet little is known about this protein in lung adenocarcinoma (LAC). The aim of this study was to evaluate the prognostic value of cripto-1 expression in a cohort of patients with LAC. Tumours from 290 patients with pathologically confirmed LAC were used for an immunohistochemical analysis of cripto-1 expression. The correlation between cripto-1 expression and the clinicopathological parameters of patients, EGFR-TKI sensitivity was analysed. Significant associations between cripto-1 expression and pT status, pN status, pTNM status, E-cadherin expression and EGFR-TKI sensitivity were identified. Compared with patients with low cripto-1 expression, patients with high cripto-1 expression exhibited significantly poorer progression-free survival (PFS) and overall survival (OS). Moreover, multivariate analyses showed that high cripto-1 expression was an independent predictor of worse survival of patients with LAC. The combination of cripto-1 expression and serum CEA level was correlated with both PFS and OS. In conclusion, cripto-1 may be a potential prognostic biomarker of survival in patients with LAC.
Collapse
|
21
|
Aykul S, Parenti A, Chu KY, Reske J, Floer M, Ralston A, Martinez-Hackert E. Biochemical and Cellular Analysis Reveals Ligand Binding Specificities, a Molecular Basis for Ligand Recognition, and Membrane Association-dependent Activities of Cripto-1 and Cryptic. J Biol Chem 2017; 292:4138-4151. [PMID: 28126904 PMCID: PMC5354514 DOI: 10.1074/jbc.m116.747501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/25/2017] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor β (TGF-β) pathways are key determinants of cell fate in animals. Their basic mechanism of action is simple. However, to produce cell-specific responses, TGF-β pathways are heavily regulated by secondary factors, such as membrane-associated EGF-CFC family proteins. Cellular activities of EGF-CFC proteins have been described, but their molecular functions, including how the mammalian homologs Cripto-1 and Cryptic recognize and regulate TGF-β family ligands, are less clear. Here we use purified human Cripto-1 and mouse Cryptic produced in mammalian cells to show that these two EGF-CFC homologs have distinct, highly specific ligand binding activities. Cripto-1 interacts with BMP-4 in addition to its known partner Nodal, whereas Cryptic interacts only with Activin B. These interactions depend on the integrity of the protein, as truncated or deglycosylated Cripto-1 lacked BMP-4 binding activity. Significantly, Cripto-1 and Cryptic blocked binding of their cognate ligands to type I and type II TGF-β receptors, indicating that Cripto-1 and Cryptic contact ligands at their receptor interaction surfaces and, thus, that they could inhibit their ligands. Indeed, soluble Cripto-1 and Cryptic inhibited ligand signaling in various cell-based assays, including SMAD-mediated luciferase reporter gene expression, and differentiation of a multipotent stem cell line. But in agreement with previous work, the membrane bound form of Cripto-1 potentiated signaling, revealing a critical role of membrane association for its established cellular activity. Thus, our studies provide new insights into the mechanism of ligand recognition by this enigmatic family of membrane-anchored TGF-β family signaling regulators and link membrane association with their signal potentiating activities.
Collapse
Affiliation(s)
- Senem Aykul
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Anthony Parenti
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Kit Yee Chu
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Jake Reske
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Monique Floer
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Amy Ralston
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| | - Erik Martinez-Hackert
- From the Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824-1319
| |
Collapse
|
22
|
Bodenstine TM, Chandler GS, Seftor REB, Seftor EA, Hendrix MJC. Plasticity underlies tumor progression: role of Nodal signaling. Cancer Metastasis Rev 2016; 35:21-39. [PMID: 26951550 DOI: 10.1007/s10555-016-9605-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The transforming growth factor beta (TGFβ) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFβ family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Thomas M Bodenstine
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Grace S Chandler
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Richard E B Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Elisabeth A Seftor
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA
| | - Mary J C Hendrix
- Stanley Manne Children's Research Institute, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 225 E. Chicago Avenue, Box 222, Chicago, IL, 60611, USA.
| |
Collapse
|
23
|
Xu CH, Wang Y, Qian LH, Yu LK, Zhang XW, Wang QB. Serum Cripto-1 is a novel biomarker for non-small cell lung cancer diagnosis and prognosis. CLINICAL RESPIRATORY JOURNAL 2016; 11:765-771. [PMID: 26605871 DOI: 10.1111/crj.12414] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/27/2015] [Accepted: 11/13/2015] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Cripto-1 (CR-1) is highly expressed in several different types of human tumors. However, the clinical significance of CR-1 expression in serum specimens from non-small cell lung cancer (NSCLC) patients has not yet been determined. OBJECTIVES The aim of this study was to explore the diagnostic and prognostic value of serum CR-1 levels in patients with NSCLC. METHODS Serum specimens from 592 NSCLC patients, 180 benign lung disease patients and 240 healthy controls were collected. The concentrations of CR-1 were measured by sandwich enzyme-linked immunosorbent assay. RESULTS Patients with NSCLC had higher serum CR-1 levels than the controls (P < 0.01) and patients with benign lung diseases (P < 0.01). When a cutoff point of 1.8 ng/mL was selected (diagnostic specificity 95%), the diagnostic sensitivity for NSCLC is 56.8%. About 37.5% of carcinoembryonic antigen (CEA)-negative lung cancer patients were CR-1 positive at 95% specificity. In patients with stage I/II lung cancer, use of these two markers in combination results in almost 21% increase in sensitivity, at 95% specificity, compared with CEA alone. Uni-variate analysis revealed that NSCLC patients with positive CR-1 had a shorter overall survival (OS) and progression-free survival (PFS) than those with negative CR-1 [hazard ratio (HR) of 2.93, P = 0.005; HR of 2.12, P = 0.005]. Cox multi-variate analysis indicated that CR-1 was an independent prognostic indicator of PFS and OS (HR of 1.91, P = 0.006; HR of 1.82, P = 0.007). Kaplan-Meier survival curves further confirmed that patients with negative CR-1 had longer PFS and OS (P = 0.026 and P = 0.011, respectively). CONCLUSIONS In conclusion, measurement of serum CR-1 is a useful diagnostic and prognostic marker for NSCLC patients.
Collapse
Affiliation(s)
- Chun Hua Xu
- Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing, China.,Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing, China
| | - Yan Wang
- Department of Radiology, Nanjing Chest Hospital, Nanjing, China
| | - Li Hua Qian
- Department of Respiratory Medicine, Nanjing Pukou Central Hospital, Nanjing, China
| | - Li Ke Yu
- Department of Respiratory Medicine, Nanjing Chest Hospital, Nanjing, China.,Clinical Center of Nanjing Respiratory Diseases and Imaging, Nanjing, China
| | - Xiu Wei Zhang
- Department of Respiratory Medicine, Nanjing Jiangning Hospital, Nanjing, China
| | - Qing Bo Wang
- Department of Geriatrics Medicine, Nanjing Second Hospital, Nanjing, China
| |
Collapse
|
24
|
Bao D, Lu D, Liu N, Dong W, Lu YD, Qin C, Zhang LF. Tomoregulin-1 prevents cardiac hypertrophy after pressure overload in mice by inhibiting TAK1-JNK pathways. Dis Model Mech 2015; 8:795-804. [PMID: 26092120 PMCID: PMC4527297 DOI: 10.1242/dmm.021303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/09/2015] [Indexed: 11/20/2022] Open
Abstract
Cardiac hypertrophy is associated with many forms of heart disease, and identifying important modifier genes involved in the pathogenesis of cardiac hypertrophy could lead to the development of new therapeutic strategies. Tomoregulin-1 is a growth factor that is primarily involved in embryonic development and adult central nervous system (CNS) function, and it is expressed abnormally in a variety of CNS pathologies. Tomoregulin-1 is also expressed in the myocardium. However, the effects of tomoregulin-1 on the heart, particularly on cardiac hypertrophy, remains unknown. The aim of the study is to examine whether and by what mechanism tomoregulin-1 regulates the development of cardiac hypertrophy induced by pressure overload. In this study, we found that tomoregulin-1 was significantly upregulated in two cardiac hypertrophy models: cTnT(R92Q) transgenic mice and thoracic aorta constriction (TAC)-induced cardiac hypertrophy mice. The transgenic overexpression of tomoregulin-1 increased the survival rate, improved the cardiac geometry and functional parameters of echocardiography, and decreased the degree of cardiac hypertrophy of the TAC mice, whereas knockdown of tomoregulin-1 expression resulted in an opposite phenotype and exacerbated phenotypes of cardiac hypertrophy induced by TAC. A possible mechanism by which tomoregulin-1 regulates the development of cardiac hypertrophy in TAC-induced cardiac hypertrophy is through inhibiting TGFβ non-canonical (TAK1-JNK) pathways in the myocardium. Tomoregulin-1 plays a protective role in the modulation of adverse cardiac remodeling from pressure overload in mice. Tomoregulin-1 could be a therapeutic target to control the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Dan Bao
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Ning Liu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Ying-Dong Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Chuan Qin
- Key Laboratory of Human Disease Animal Model, State Administration of Traditional Chinese Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
25
|
Tumorigenic factor CRIPTO-1 is immunolocalized in extravillous cytotrophoblast in placenta creta. BIOMED RESEARCH INTERNATIONAL 2014; 2014:892856. [PMID: 25165718 PMCID: PMC4140153 DOI: 10.1155/2014/892856] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/10/2014] [Indexed: 12/21/2022]
Abstract
CRIPTO-(CR)1 is a protein associated with tumorigenesis and metastasis. Here we demonstrate that CR-1 expression in normal and creta placentas is associated with various degrees of uterine invasion. Cytokeratin (CK) and CR-1 protein expression was visualized by immunohistochemical staining of formalin-fixed, paraffin-embedded placental specimens (control placentas, n = 9; accreta, n = 6; increta, n = 10; percreta, n = 15). The pattern of extravillous trophoblast (EVT) cell morphology was distinctive in creta placentas: densely-compacted cell columns and large star-shaped cells with a typically migratory phenotype, not common among third trimester control placentas. Quantification revealed higher CR-1 immunoreactivities in accreta (P = 0.001), increta (P = 0.0002), and percreta placentas (P = 0.001) than in controls. In contrast to controls, there was a significant positive relationship between CR-1 and CK reactivity in all creta placentas (accreta, P = 0.02; increta, P = 0.0001, and percreta, P = 0.025). This study demonstrated CR-1 expression in the placental bed, its increased expression in creta placentas, and EVT cells as the main CR-1-producing cell type. Morphological examination revealed an immature and invasive trophoblast profile in creta placentas, suggesting impairment of the trophoblast differentiation pathway. These findings provide important new insights into the pathophysiology of abnormal creta placentation and its gestational consequences.
Collapse
|
26
|
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: 0.8] [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
|
27
|
Pilgaard L, Mortensen JH, Henriksen M, Olesen P, Sørensen P, Laursen R, Vyberg M, Agger R, Zachar V, Moos T, Duroux M. Cripto-1 expression in glioblastoma multiforme. Brain Pathol 2014; 24:360-70. [PMID: 24521322 DOI: 10.1111/bpa.12131] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/05/2014] [Indexed: 01/24/2023] Open
Abstract
Human glioblastoma multiforme (GBM) is an aggressive cancer with a very poor prognosis. Cripto-1 (CR-1) has a key regulatory role in embryogenesis, while in adult tissue re-expression of CR-1 has been correlated to malignant progression in solid cancers of non-neuronal origin. As CR-1 expression has yet to be described in cerebral cancer and CR-1 is regulated by signaling pathways dysregulated in GBM, we aimed to investigate CR-1 in the context of expression in GBM. The study was performed using enzyme-linked immunosorbent assay (ELISA), Western blotting, polymerase chain reaction (PCR) and immunohistochemistry to analyze the blood and tissue from 28 GBM and 4 low-grade glioma patients. Within the patient cohort, we found high CR-1 protein levels in blood plasma to significantly correlate with a shorter overall survival. We identified CR-1 in different areas of GBM tissue, including perivascular tumor cells, and in endothelial cells. Collectively, our data suggest that CR-1 could be a prognostic biomarker for GBM with the potential of being a therapeutic target.
Collapse
Affiliation(s)
- Linda Pilgaard
- Laboratory of Cancer Biology, Biomedicine Group, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hu XF, Yang E, Li J, Xing PX. MUC1 cytoplasmic tail: a potential therapeutic target for ovarian carcinoma. Expert Rev Anticancer Ther 2014; 6:1261-71. [PMID: 16925492 DOI: 10.1586/14737140.6.8.1261] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ovarian cancer is often a lethal disease, since the occult progression of the tumor within the peritoneal cavity results in late diagnosis and treatment failure. The identification of molecular events specific to metastasis is critical for the development of effective therapies. MUC1 is aberrantly overexpressed by most ovarian cancer and regarded as a molecular target for ovarian cancer. This review focuses on the latest advances regarding a signaling region in the MUC1 C-terminal subunit-mediated c-Src signaling pathways in malignant transformation, invasion and metastasis. Disruption of MUC1-C-terminal subunit-associated c-Src signaling by targeting the specific sites might represent a novel immunotherapeutic approach for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Xiu Feng Hu
- Cancer Immunotherapy Laboratory, Burnet Institute Incorporating Austin Research Institute, Studley Road, Heidelberg, Victoria 3084, Australia.
| | | | | | | |
Collapse
|
29
|
Stolfi C, Marafini I, De Simone V, Pallone F, Monteleone G. The dual role of Smad7 in the control of cancer growth and metastasis. Int J Mol Sci 2013; 14:23774-90. [PMID: 24317436 PMCID: PMC3876077 DOI: 10.3390/ijms141223774] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 02/07/2023] Open
Abstract
Smad7 was initially identified as an inhibitor of Transforming growth factor (TGF)-β due mainly to its ability to bind TGF-β receptor type I and prevent TGF-β-associated Smad signaling. More recently, it has been demonstrated that Smad7 can interact with other intracellular proteins and regulate also TGF-β-independent signaling pathways thus making a valid contribution to the neoplastic processes in various organs. In particular, data emerging from experimental studies indicate that Smad7 may differently modulate the course of various tumors depending on the context analyzed. These observations, together with the demonstration that Smad7 expression is deregulated in many cancers, suggest that therapeutic interventions around Smad7 can help interfere with the development/progression of human cancers. In this article we review and discuss the available data supporting the role of Smad7 in the modulation of cancer growth and progression.
Collapse
Affiliation(s)
- Carmine Stolfi
- Authors to whom correspondence should be addressed; E-Mails: (C.S.); (G.M.); Tel.: +39-6-7259-6150 (G.S.); Fax: +39-6-7259-6391 (G.S.)
| | | | | | | | - Giovanni Monteleone
- Authors to whom correspondence should be addressed; E-Mails: (C.S.); (G.M.); Tel.: +39-6-7259-6150 (G.S.); Fax: +39-6-7259-6391 (G.S.)
| |
Collapse
|
30
|
Frasca F, Vella V, Nicolosi ML, Messina RL, Gianì F, Lotta S, Vigneri P, Regalbuto C, Vigneri R. Thyroid cancer cell resistance to gefitinib depends on the constitutive oncogenic activation of the ERK pathway. J Clin Endocrinol Metab 2013; 98:2502-12. [PMID: 23559083 DOI: 10.1210/jc.2012-3623] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CONTEXT Poorly differentiated thyroid carcinomas are refractory to common anticancer therapies, and novel inhibitors are being tested in these deadly malignancies. The epidermal growth factor receptor (EGFR) tyrosine kinase represents an attractive target for treatment because it is up-regulated in thyroid cancer and plays a role in cancer progression. However, EGFR inhibitors have provided poor results in thyroid carcinomas. OBJECTIVE We evaluated the possible mechanism underlying the resistance of thyroid cancer cells to EGFR inhibitors. DESIGN We tested the effect of the EGFR tyrosine kinase inhibitor gefitinib in a panel of thyroid cancer cell lines. RESULTS We found that in most of the cell lines, although gefitinib inhibited EGFR phosphorylation, it was poorly effective in reducing cell viability. gefitinib, however, was able to inhibit epidermal growth factor-induced cell migration and matrix invasion. In most thyroid cancer cell lines, gefitinib significantly inhibited Akt phosphorylation by inhibiting EGFR activation, but it had limited or no effect on ERK phosphorylation. The poor cell response to gefitinib was associated with genetic alterations, leading to constitutive activation of the ERK pathway, including BRAF(V600E) and HRAS(G12A/Q61R) mutations and RET/PTC1 rearrangement. When BRAF(V600E)-positive thyroid cancer cells were incubated with the specific BRAF inhibitor PLX4032, sensitivity to gefitinib was restored. Similar results were obtained with rat sarcoma and RET/papillary thyroid cancer inhibitors. CONCLUSIONS These results indicate that thyroid cancer resistance to gefitinib is due to the constitutive activation of the mitogenic pathway by either signals downstream of EGFR or other tyrosine kinase receptors. This resistance can be overcome by the combined use of selective inhibitors.
Collapse
Affiliation(s)
- Francesco Frasca
- Endocrinology Unit, Department of Clinical and Molecular Bio-Medicine, University of Catania, Garibaldi-Nesima Medical Center, Via Palermo 636, 95122 Catania, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Behrens AN, Ren Y, Ferdous A, Garry DJ, Martin CM. Nkx2-5 Regulates Tdgf1 (Cripto) Early During Cardiac Development. ACTA ACUST UNITED AC 2013; Suppl 11:1-4. [PMID: 24069547 DOI: 10.4172/2155-9880.s11-003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Congenital Heart Disease (CHD) is the most frequent and deadly birth defect. Patients with CHD that survive the neonatal period often progress to develop advanced heart failure requiring specialized treatment including cardiac transplantation. A full understanding of the transcriptional networks that direct cardiac progenitors during heart development will enhance our understanding of both normal cardiac function and pathological states. These findings will also have important applications for emerging therapies and the treatment of congenital heart disease. Furthermore, a number of shared transcriptional pathways or networks have been proposed to regulate the development and regeneration of tissues such as the heart. We have utilized transgenic technology to isolate and characterize cardiac progenitor cells from the developing mouse heart and have begun to define specific transcriptional networks of cardiovascular development. Initial studies identified Tdgf1 as a potential target of Nkx2-5. To mechanistically dissect the regulation of this molecular program, we utilized an array of molecular biological techniques to confirm that Nkx2-5 is an upstream regulator of the Tdgf1 gene in early cardiac development. These studies further define Nkx2-5 mediated transcriptional networks and enhance our understanding of cardiac morphogenesis.
Collapse
Affiliation(s)
- Ann N Behrens
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | |
Collapse
|
32
|
Miharada K, Karlsson G, Rehn M, Rörby E, Siva K, Cammenga J, Karlsson S. Hematopoietic stem cells are regulated by Cripto, as an intermediary of HIF-1α in the hypoxic bone marrow niche. Ann N Y Acad Sci 2012; 1266:55-62. [PMID: 22901256 DOI: 10.1111/j.1749-6632.2012.06564.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cripto has been known as an embryonic stem (ES)- or tumor-related soluble/cell membrane protein. In this study, we demonstrated that Cripto has a role as an important regulatory factor for hematopoietic stem cells (HSCs). Recombinant Cripto sustained the reconstitution ability of HSCs in vitro. Flow cytometry analysis uncovered that GRP78, one of the candidate receptors for Cripto, was expressed on a subset of HSCs and could distinguish dormant/myeloid-biased HSCs and active/lymphoid-biased HSCs. Cripto is expressed in hypoxic endosteal niche cells where GRP78(+) HSCs mainly reside. Proteomics analysis revealed that Cripto-GRP78 binding stimulates glycolytic metabolism-related proteins and results in lower mitochondrial potential in HSCs. Furthermore, conditional knockout mice for HIF-1α, a master regulator of hypoxic responses, showed reduced Cripto expression and decreased GRP78(+) HSCs in the endosteal niche area. Thus, Cripto-GRP78 is a novel HSC regulatory signal mainly working in the hypoxic niche.
Collapse
Affiliation(s)
- Kenichi Miharada
- Department for Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
33
|
Gray PC, Vale W. Cripto/GRP78 modulation of the TGF-β pathway in development and oncogenesis. FEBS Lett 2012; 586:1836-45. [PMID: 22306319 PMCID: PMC3723343 DOI: 10.1016/j.febslet.2012.01.051] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 10/14/2022]
Abstract
Cripto is a small, GPI-anchored signaling protein that regulates cellular survival, proliferation, differentiation and migration during normal developmental processes and tumorigenesis. Cripto functions as an obligatory co-receptor for the TGF-β ligands Nodal, GDF1 and GDF3 but attenuates signaling of others such as activin-A, activin-B and TGF-β1. Soluble, secreted forms of Cripto also activate Src, ras/raf/MAPK and PI3K/Akt pathways via a mechanism that remains largely obscure. This review describes the biological roles and signaling mechanisms of Cripto, highlighting our identification of the 78 kDa glucose regulated protein (GRP78) as a cell surface receptor/co-factor required for Cripto signaling via both TGF-β and Src/MAPK/PI3K pathways. We discuss emerging evidence indicating that Cripto/GRP78 signaling regulates normal somatic stem cells and their tumorigenic counterparts.
Collapse
Affiliation(s)
- Peter C Gray
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, United States.
| | | |
Collapse
|
34
|
Miharada K, Karlsson G, Rehn M, Rörby E, Siva K, Cammenga J, Karlsson S. Cripto regulates hematopoietic stem cells as a hypoxic-niche-related factor through cell surface receptor GRP78. Cell Stem Cell 2012; 9:330-44. [PMID: 21982233 DOI: 10.1016/j.stem.2011.07.016] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/14/2011] [Accepted: 07/29/2011] [Indexed: 02/06/2023]
Abstract
Hematopoietic stem cells (HSCs) are maintained in hypoxic niches in endosteal regions of bones. Here we demonstrate that Cripto and its receptor GRP78 are important regulators of HSCs in the niche. Flow cytometry analyses revealed two distinct subpopulations of CD34(-)KSL cells based on the expression of GRP78, and these populations showed different reconstitution potential in transplantation assays. GRP78(+)HSCs mainly reside in the endosteal area, are more hypoxic, and exhibit a lower mitochondrial potential, and their HSC capacity was maintained in vitro by Cripto through induction of higher glycolytic activity. Additionally, HIF-1α KO mice have decreased numbers of GRP78(+)HSCs and reduced expression of Cripto in the endosteal niche. Furthermore, blocking GRP78 induced a movement of HSCs from the endosteal to the central marrow area. These data suggest that Cripto/GRP78 signaling is an important pathway that regulates HSC quiescence and maintains HSCs in hypoxia as an intermediary of HIF-1α.
Collapse
Affiliation(s)
- Kenichi Miharada
- Department for Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, Sweden.
| | | | | | | | | | | | | |
Collapse
|
35
|
Okada S, Ochiai H, Saito A, Sato Y, Azuma T. Regulation of Nodal-Lefty Expression by TGF-^|^beta; Induced Pluripotent Stem (iPS) Cells Derived from Mouse Oral Mucosal Tissue. J HARD TISSUE BIOL 2012. [DOI: 10.2485/jhtb.21.391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
36
|
Yoon HJ, Hong JS, Shin WJ, Lee YJ, Hong KO, Lee JI, Hong SP, Hong SD. The role of Cripto-1 in the tumorigenesis and progression of oral squamous cell carcinoma. Oral Oncol 2011; 47:1023-31. [PMID: 21824804 DOI: 10.1016/j.oraloncology.2011.07.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/20/2011] [Accepted: 07/20/2011] [Indexed: 11/17/2022]
Abstract
Oral squamous cell carcinoma (OSCC), the most common malignancy of the oral cavity, remains a lethal disease in over 50% of cases diagnosed annually, due mostly to late detection of this cancer in its advanced stages despite the easy accessibility of the oral cavity for regular examinations. Cripto-1 is a member of the epidermal growth factor (EGF)-CFC protein family and is involved in the activation of several different signaling pathways during embryonic development and cellular transformation. Although the Cripto-1 protein is overexpressed in several human cancers including breast, colon, cervix, gastric, and pancreatic cancer, no prior study has evaluated Cripto-1 expression in OSCC. Therefore, our aims in this study were to examine Cripto-1 expression in clinical samples of OSCC patients using immunohistochemistry, to analyze the correlation between Cripto-1 expression and clinicopathologic parameters, and to identify the oncogenic roles of Cripto-1 in OSCC cell lines. Both epithelial dysplasia (73.3%) and OSCC (55.5%) tissue samples showed significantly higher expression of Cripto-1 than normal mucosa (20%) (p=0.031). In the OSCC samples, there was a significant correlation between Cripto-1 expression and the histological differentiation of OSCC (p=0.015) and a high PCNA index (p=0.011). The in vitro cell proliferation assays demonstrated that recombinant human Cripto-1 (rhCripto-1) induced both SCC-4 and SCC-25 cells to proliferate as compared with control cells (p<0.05 and p<0.01, respectively). In in vitro migration assays, treatment of SCC-4 and SCC-25 cells with rhCripto-1 protein induced a 2.4-fold and 1.7-fold-increase in cell migration, respectively (p=0.000 and p=0.008, respectively). Taken together, our data suggest that Cripto-1 plays a role in the malignant transformation of the oral mucosa and is involved in the tumorigenesis and progression of OSCC by promoting the growth and migration of malignant cells.
Collapse
Affiliation(s)
- Hye-Jung Yoon
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, 28 Yeongeon-dong, Chongno-gu, Seoul 110-749, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Lawrence MG, Margaryan NV, Loessner D, Collins A, Kerr KM, Turner M, Seftor EA, Stephens CR, Lai J, BioResource APC, Postovit LM, Clements JA, Hendrix MJ. Reactivation of embryonic nodal signaling is associated with tumor progression and promotes the growth of prostate cancer cells. Prostate 2011; 71:1198-209. [PMID: 21656830 PMCID: PMC3234312 DOI: 10.1002/pros.21335] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 12/07/2010] [Indexed: 11/06/2022]
Abstract
BACKGROUND Nodal is a member of the transforming growth factor β (TGFβ) superfamily that directs embryonic patterning and promotes the plasticity and tumorigenicity of tumor cells, but its role in the prostate is unknown. The goal of this study was to characterize the expression and function of Nodal in prostate cancer and determine whether, like other TGFβ ligands, it modulates androgen receptor (AR) activity. METHODS Nodal expression was investigated using immunohistochemistry of tissue microarrays and Western blots of prostate cell lines. The functional role of Nodal was examined using Matrigel and soft agar growth assays. Cross-talk between Nodal and AR signaling was assessed with luciferase reporter assays and expression of endogenous androgen regulated genes. RESULTS Significantly increased Nodal expression was observed in cancer compared with benign prostate specimens. Nodal was only expressed by DU145 and PC3 cells. All cell lines expressed Nodal's co-receptor, Cripto-1, but lacked Lefty, a critical negative regulator of Nodal signaling. Recombinant human Nodal triggered downstream Smad2 phosphorylation in DU145 and LNCaP cells, and stable transfection of pre-pro-Nodal enhanced the growth of LNCaP cells in Matrigel and soft agar. Finally, Nodal attenuated AR signaling, reducing the activity of a PSA promoter construct in luciferase assays and down-regulating the endogenous expression of androgen regulated genes. CONCLUSIONS An aberrant Nodal signaling pathway is re-expressed and functionally active in prostate cancer cells.
Collapse
Affiliation(s)
- Mitchell G. Lawrence
- Australian Prostate Cancer Research Centre-Queensland and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, 4059
| | - Naira V. Margaryan
- Program in Cancer Biology and Epigenomics, Children’s Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA, 60614
| | - Daniela Loessner
- Australian Prostate Cancer Research Centre-Queensland and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, 4059
| | - Angus Collins
- Sullivan Nicolaides Pathology, Brisbane, Queensland, Australia, 4068
| | - Kris M. Kerr
- Sullivan Nicolaides Pathology, Brisbane, Queensland, Australia, 4068
| | - Megan Turner
- Sullivan Nicolaides Pathology, Brisbane, Queensland, Australia, 4068
| | - Elisabeth A. Seftor
- Program in Cancer Biology and Epigenomics, Children’s Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA, 60614
| | - Carson R. Stephens
- Australian Prostate Cancer Research Centre-Queensland and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, 4059
| | - John Lai
- Australian Prostate Cancer Research Centre-Queensland and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, 4059
| | | | - Lynne-Marie Postovit
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | - Judith A. Clements
- Australian Prostate Cancer Research Centre-Queensland and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, 4059
- Correspondence: Mary J.C. Hendrix, Children’s Memorial Research Center, Northwestern University, Feinberg School of Medicine, 2300 Children’s Plaza, Box 222, Chicago, IL 60614-3394, and Judith A. Clements, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland, 4059,
| | - Mary J.C. Hendrix
- Program in Cancer Biology and Epigenomics, Children’s Memorial Research Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA, 60614
- Correspondence: Mary J.C. Hendrix, Children’s Memorial Research Center, Northwestern University, Feinberg School of Medicine, 2300 Children’s Plaza, Box 222, Chicago, IL 60614-3394, and Judith A. Clements, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland, 4059,
| |
Collapse
|
38
|
Ravisankar V, Singh TP, Manoj N. Molecular evolution of the EGF-CFC protein family. Gene 2011; 482:43-50. [PMID: 21640172 DOI: 10.1016/j.gene.2011.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/16/2011] [Indexed: 10/18/2022]
Abstract
The epidermal growth factor-Cripto-1/FRL-1/Cryptic (EGF-CFC) proteins, characterized by the highly conserved EGF and CFC domains, are extracellular membrane associated growth factor-like glycoproteins. These proteins are essential components of the Nodal signaling pathway during early vertebrate embryogenesis. Homologs of the EGF-CFC family have also been implicated in tumorigenesis in humans. Yet, little is known about the mode of molecular evolution in this family. Here we investigate the origin, extent of conservation and evolutionary relationships of EGF-CFC proteins across the metazoa. The results suggest that the first appearance of the EGF-CFC gene occurred in the ancestor of the deuterostomes. Phylogenetic analysis supports the classification of the family into distinct subfamilies that appear to have evolved through lineage-specific duplication and divergence. Site-specific analyses of evolutionary rate shifts between the two major mammalian paralogous subfamilies, Cripto and Cryptic, reveal critical amino acid sites that may account for the observed functional divergence. Furthermore, estimates of functional divergence suggest that rapid change of evolutionary rates at sites located mainly in the CFC domain may contribute towards distinct functional properties of the two paralogs.
Collapse
Affiliation(s)
- V Ravisankar
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, India.
| | | | | |
Collapse
|
39
|
Shi Y, Bao YL, Wu Y, Yu CL, Huang YX, Sun Y, Zheng LH, Li YX. Alantolactone Inhibits Cell Proliferation by Interrupting the Interaction between Cripto-1 and Activin Receptor Type II A in Activin Signaling Pathway. ACTA ACUST UNITED AC 2011; 16:525-35. [DOI: 10.1177/1087057111398486] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
It has been suggested that deregulation of activin signaling contributes to tumor formation. Activin signaling is blocked in cancer cells due to the complex formed by Cripto-1, activin, and activin receptor type II (ActRII). In this study, the authors used a mammalian two-hybrid system to construct a drug screening model to obtain a small molecular inhibitor capable of interrupting the interaction between Cripto-1 and ActRII. They screened 300 natural components and identified alantolactone. Data suggested that alantolactone induced activin/SMAD3 signaling in human colon adenocarcinoma HCT-8 cells. The authors also found that alantolactone exhibited antiproliferative function specific to tumor cells, with almost no toxicity to normal cells at a concentration of 5 µg/mL. Furthermore, they proved that the antiproliferative function of alantolactone was activin/SMAD3 dependent. These results suggest that alantolactone performs its antitumor effect by interrupting the interaction between Cripto-1 and the activin receptor type IIA in the activin signaling pathway. Moreover, screening for inhibitors of Cripto-1/ActRII is a potentially beneficial approach to aid in discovering novel cancer treatment.
Collapse
Affiliation(s)
- Ying Shi
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China, Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yong Li Bao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China, Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yin Wu
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Chun Lei Yu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China, Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yan Xin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Ying Sun
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China
| | - Li Hua Zheng
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China, Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China
| | - Yu Xin Li
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China, Institute of Genetics and Cytology, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
40
|
Ciarmela P, Bloise E, Gray PC, Carrarelli P, Islam MS, De Pascalis F, Severi FM, Vale W, Castellucci M, Petraglia F. Activin-A and myostatin response and steroid regulation in human myometrium: disruption of their signalling in uterine fibroid. J Clin Endocrinol Metab 2011; 96:755-65. [PMID: 21177794 PMCID: PMC3047220 DOI: 10.1210/jc.2010-0501] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Investigation of activin-A (A) and myostatin (M) in human myometrium (HM) and leiomyoma (HL) will explain their involvement in human myometrial pathophysiology. OBJECTIVE We aimed to investigate A and M response and steroid regulation in HM. We also evaluated A and M expression and response in HL. DESIGN Tissues were analyzed and cultured. PATIENTS Patients included fertile (in proliferative phase) and menopausal women undergoing hysterectomy. INTERVENTIONS HM explant cultures were treated with A and M (for Smad-7 mRNA quantification) or estrogen and progesterone (for A and M mRNA quantification). A and M expression levels were also evaluated in menopausal (physiological absence of steroids) HM specimens. A and M and their receptors were evaluated in HL (n = 8, diameter 5-8 cm) compared with their matched HM. HL explants cultures were treated with A and M (for Smad7 mRNA quantification), and, to explain the absence of response, the levels of follistatin, follistatin-related gene (FLRG), and Cripto were evaluated. RESULTS A and M increased Smad7 expression in HM explants. A and M mRNAs were both reduced after estradiol treatment, unchanged after progesterone treatment, but were higher in menopausal than fertile (in proliferative phase) specimens. A, M, and FLRG were expressed at higher levels in HL compared with adjacent HM, whereas the receptors, follistatin, and Smad7 mRNAs resulted unchanged. Cripto mRNA was expressed only in HL. CONCLUSIONS A and M act on human HM and are regulated by steroids. In HL there is an increase of A, M, FLRG, and Cripto expression.
Collapse
Affiliation(s)
- Pasquapina Ciarmela
- Department of Molecular Pathology and Innovative Therapies, Faculty of Medicine, Polytechnic University of Marche, via Tronto 10/a, 60020 Ancona, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Watanabe K, Meyer MJ, Strizzi L, Lee JM, Gonzales M, Bianco C, Nagaoka T, Farid SS, Margaryan N, Hendrix MJC, Vonderhaar BK, Salomon DS. Cripto-1 is a cell surface marker for a tumorigenic, undifferentiated subpopulation in human embryonal carcinoma cells. Stem Cells 2011; 28:1303-14. [PMID: 20549704 DOI: 10.1002/stem.463] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Deregulation of stem cells is associated with the generation and progression of malignant tumors. In addition, genes that are associated with early embryogenesis are frequently expressed in cancer. Cripto-1 (CR-1), a glycosylphosphatidylinositol-linked glycoprotein, is expressed during early embryogenesis and in various human carcinomas. We demonstrated that human embryonal carcinoma (EC) cells are heterogeneous for CR-1 expression and consist of two distinct subpopulations: a CR-1(High) and a CR-1(Low) population. By segregating CR-1(High) and CR-1(Low) populations of NTERA2/D1 EC cells by fluorescence-activated cell sorting, we demonstrated that CR-1(High) cells were more tumorigenic than CR-1(Low) cells by an in vitro tumor sphere assay and by in vivo xenograft formation. The CR-1(High) population was enriched in mRNA expression for the pluripotent embryonic stem (ES) cell genes Oct4, Sox2, and Nanog. CR-1 expression in NTERA2/D1 cells was regulated by a Smad2/3-dependent autocrine loop, by the ES cell-related transcription factors Oct4/Nanog, and partially by the DNA methylation status of the promoter region. These results demonstrate that CR-1 expression is enriched in an undifferentiated, tumorigenic subpopulation and is regulated by key regulators of pluripotent stem cells.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- Mammary Biology and Tumorigenesis Laboratory, Robert H. LurieCancer Center Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Watanabe K, Salomon DS. Intercellular transfer regulation of the paracrine activity of GPI-anchored Cripto-1 as a Nodal co-receptor. Biochem Biophys Res Commun 2010; 403:108-13. [PMID: 21055389 DOI: 10.1016/j.bbrc.2010.10.128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 10/27/2010] [Indexed: 01/15/2023]
Abstract
Cripto-1 (CR-1) is a glycosylphosphatidylinositol-anchored glycoprotein which acts as an obligate co-receptor of a TGFβ family ligand, Nodal. Previous studies have demonstrated that CR-1 functions in a paracrine fashion by a cellular mechanism which has not been fully described. This paracrine activity was observed only when CR-1 was expressed as a membrane-bound form and was abolished when CR-1 was expressed in a soluble form. In the current study, we found that there were few biochemical differences in post-translational modifications between membrane-anchored and soluble forms of CR-1. Flow cytometric analysis revealed an intercellular transfer of the membrane-bound form of CR-1 between cells. CR-1-expressing cells formed unique membrane extensions, generated more membrane fragments than control cells, and exhibited enhanced cellular adhesion. Thus, expression of CR-1 may alter the physiochemical properties of the plasma membrane resulting in an enhancement of intercellular transfer of cellular signaling components which may account for the paracrine activity of CR-1.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
43
|
Calvanese L, Marasco D, Doti N, Saporito A, D'Auria G, Paolillo L, Ruvo M, Falcigno L. Structural investigations on the Nodal-Cripto binding: A theoretical and experimental approach. Biopolymers 2010; 93:1011-21. [DOI: 10.1002/bip.21517] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Micalizzi DS, Farabaugh SM, Ford HL. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia 2010; 15:117-34. [PMID: 20490631 PMCID: PMC2886089 DOI: 10.1007/s10911-010-9178-9] [Citation(s) in RCA: 739] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 04/26/2010] [Indexed: 02/07/2023] Open
Abstract
From the earliest stages of embryonic development, cells of epithelial and mesenchymal origin contribute to the structure and function of developing organs. However, these phenotypes are not always permanent, and instead, under the appropriate conditions, epithelial and mesenchymal cells convert between these two phenotypes. These processes, termed Epithelial-Mesenchymal Transition (EMT), or the reverse Mesenchymal-Epithelial Transition (MET), are required for complex body patterning and morphogenesis. In addition, epithelial plasticity and the acquisition of invasive properties without the full commitment to a mesenchymal phenotype are critical in development, particularly during branching morphogenesis in the mammary gland. Recent work in cancer has identified an analogous plasticity of cellular phenotypes whereby epithelial cancer cells acquire mesenchymal features that permit escape from the primary tumor. Because local invasion is thought to be a necessary first step in metastatic dissemination, EMT and epithelial plasticity are hypothesized to contribute to tumor progression. Similarities between developmental and oncogenic EMT have led to the identification of common contributing pathways, suggesting that the reactivation of developmental pathways in breast and other cancers contributes to tumor progression. For example, developmental EMT regulators including Snail/Slug, Twist, Six1, and Cripto, along with developmental signaling pathways including TGF-beta and Wnt/beta-catenin, are misexpressed in breast cancer and correlate with poor clinical outcomes. This review focuses on the parallels between epithelial plasticity/EMT in the mammary gland and other organs during development, and on a selection of developmental EMT regulators that are misexpressed specifically during breast cancer.
Collapse
Affiliation(s)
- Douglas S. Micalizzi
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045 USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Susan M. Farabaugh
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Heide L. Ford
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO 80045 USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045 USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045 USA
- Program in Molecular Biology, University of Colorado School of Medicine, Aurora, CO 80045 USA
- University of Colorado at Denver, Anschutz Medical Campus, RC1 North, Rm. 5102, Aurora, CO 80045 USA
| |
Collapse
|
45
|
Strizzi L, Postovit LM, Margaryan NV, Seftor EA, Abbott DE, Seftor REB, Salomon DS, Hendrix MJC. Emerging roles of nodal and Cripto-1: from embryogenesis to breast cancer progression. Breast Dis 2009; 29:91-103. [PMID: 19029628 DOI: 10.3233/bd-2008-29110] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Breast carcinoma cells and embryonic progenitors similarly implement stem cell-associated signaling pathways to sustain continued growth and plasticity. Indeed, recent studies have implicated signaling pathways, including those associated with the Notch, and Transforming Growth Factor-Beta (TGF-beta) superfamilies, as instrumental to both embryological development and breast cancer progression. In particular, Nodal, an embryonic morphogen belonging to the TGF-beta superfamily, and its co-receptor, Cripto-1, are requisite to both embryogenesis and mammary gland maturation. Moreover, these developmental proteins have been shown to promote breast cancer progression. Here, we review the role of Nodal and its co-receptor Cripto-1 during development and we describe how this signaling pathway may be involved in breast cancer tumorigenesis. Moreover, we emphasize the potential utility of this signaling pathway as a novel target for the treatment and diagnosis of breast cancer.
Collapse
Affiliation(s)
- Luigi Strizzi
- Children's Memorial Research Center, Cancer Biology and Epigenomics Program, Robert H. Lurie Comprehensive Cancer Center, Northwestern University's Feinberg School of Medicine, Chicago, IL 60614, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu W, Wu Z, Guan M, Lu Y. cDNA microarray analysis of pigment epithelium-derived factor-regulated gene expression profile in prostate carcinoma cells. Int J Urol 2009; 16:323-8. [PMID: 19207613 DOI: 10.1111/j.1442-2042.2008.02199.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES To clarify molecular mechanisms involved in the action of pigment epithelium-derived factor (PEDF) in hormone insensitive prostate cancer cells. METHODS Total ribonucleic acid from untreated and PEDF-treated cells was subjected to microarray analysis using BioStar 8464 microarray. Real-time polymerase chain reaction analysis was conducted to confirm the microarray data. RESULTS Twenty-seven out of 8464 genes were found altered in both cell lines. Common gene responses altered by PEDF were identified and included genes known to alter cell signaling as well as genes involved in catalytic activity, cell proliferation, angiogenesis and apoptosis. Real-time reverse transcription polymerase chain reaction, in accordance with the microarray analysis, indicated that PEDF treatment caused an upregulation in the mRNA expression level of stanniocalcin 2, brain-specific angiogenesis inhibitor 2 and growth arrest, DNA-damage-inducible, alpha, and downregulation in the messenger ribonucleic acid level of fibroblast growth factor 3, teratocarcinoma-derived growth factor, neuropilin1, and endothelial Per/ARNT/Sim domain protein1, respectively. CONCLUSIONS These findings demonstrate that PEDF administration causes significant changes in the gene expression of the prostate, providing insights into the potential role of PEDF in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
47
|
Watanabe K, Nagaoka T, Strizzi L, Mancino M, Gonzales M, Bianco C, Salomon DS. Characterization of the glycosylphosphatidylinositol-anchor signal sequence of human Cryptic with a hydrophilic extension. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1778:2671-81. [PMID: 18930707 PMCID: PMC3385650 DOI: 10.1016/j.bbamem.2008.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 09/06/2008] [Accepted: 09/12/2008] [Indexed: 10/21/2022]
Abstract
Epidermal Growth Factor-Cripto-1/FRL-1/Cryptic (EGF-CFC) proteins, including human Cripto-1 (hCFC2/hCR-1) and human Cryptic (hCFC1), are membrane-associated Nodal co-receptors, which have critical roles in vertebrate development. Most of the EGF-CFC proteins have been experimentally proven or predicted to be glycosylphosphatidylinositol (GPI)-anchored proteins. However, unlike other EGF-CFC proteins, hCFC1 does not exhibit a typical GPI-signal sequence, containing a 32-amino acid hydrophilic extension in its COOH-terminal end. Here we experimentally demonstrate that the COOH-terminal sequence of hCFC1 functions as a GPI-anchoring signal. Moreover, addition of a hydrophilic epitope tag of 55-amino acids (V5-His) after the GPI signal of hCR-1 interfered with generation of a GPI-anchored form of hCR-1. In contrast, addition of the same epitope tag to the end of GPI signal of hCFC1 did not affect the GPI-attachment of hCFC1. The COOH-terminal signal of hCFC1 could produce two different forms of the protein; a GPI-anchored form and an unprocessed form which was more prone to be secreted into the conditioned medium. The hydrophilic extension of hCFC1 negatively regulates the activity of hCFC1 as a Nodal co-receptor. These results demonstrate the presence of endogenous GPI-signal sequence with a hydrophilic extension, which can generate both GPI-anchored and soluble forms of the protein.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
| | - Tadahiro Nagaoka
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
| | - Luigi Strizzi
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
- Children’s Memorial Research Center, Robert H. Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Mario Mancino
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
- Oncology Research Center of Mercogliano (AV), Italy
| | - Monica Gonzales
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
| | - Caterina Bianco
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
| | - David S. Salomon
- Tumor Growth Factor Section, Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, MD
| |
Collapse
|
48
|
Calvanese L, Saporito A, Oliva R, D' Auria G, Pedone C, Paolillo L, Ruvo M, Marasco D, Falcigno L. Structural insights into the interaction between the Cripto CFC domain and the ALK4 receptor. J Pept Sci 2008; 15:175-83. [DOI: 10.1002/psc.1091] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
49
|
Sun C, Orozco O, Olson DL, Choi E, Garber E, Tizard R, Szak S, Sanicola M, Carulli JP. CRIPTO3, a presumed pseudogene, is expressed in cancer. Biochem Biophys Res Commun 2008; 377:215-20. [PMID: 18835250 DOI: 10.1016/j.bbrc.2008.09.113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 09/23/2008] [Indexed: 11/17/2022]
Abstract
Cripto is a cell surface protein highly expressed in certain solid tumors, and overexpression of Cripto protein is oncogenic. Cripto-1 protein is encoded by CRIPTO1 gene. CRIPTO3, a presumed pseudogene, has an open reading frame with six amino acid differences from Cripto-1. We show that CRIPTO3 mRNA is the CRIPTO message expressed in many cancer samples. A CRIPTO3 SAGE tag was found in several cancer SAGE libraries, while the CRIPTO1 tag was found in ES cell libraries. In vitro experiments indicate both Cripto-1 and Cripto-3 proteins are functional in the Nodal-dependent signal pathway. Our data indicate that CRIPTO3 is an expressed gene, particularly in certain cancers, and suggest a potentially novel mechanism of oncogenesis through activation of a retrogene.
Collapse
Affiliation(s)
- Chao Sun
- Department of Drug Discovery, Biogen Idec Inc, 14 Cambridge Center, Cambridge, MA 02142, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Blickwede M, Borlak J. Isolation and characterization of metabolically competent pulmonary epithelial cells from pig lung tissue. Xenobiotica 2008; 35:927-41. [PMID: 16393853 DOI: 10.1080/00498250500296264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Administration of drugs by inhalation opens new possibilities for entry into the systemic circulation and cultures of porcine pulmonary epithelial cells (PECs) may prove to be valuable in the prediction of pulmonary metabolism of drugs in humans. This paper, therefore, reports a method for the routine isolation and cultivation of PECs from slaughterhouse animals. On average 1.5x10(6) cells g-1 tissue were isolated by discontinuous density-gradient centrifugation. Cells were subsequently cultivated on collagen-coated plates and characterized by staining for alkaline phosphatase, by tannic acid staining of lamellar bodies and by surfactant protein (SP) expression at days 0, 3 and 6 in culture. Over 70% of purified cells were positive for SP-C and tannic acid staining and thus defined as epithelial cells of alveolar origin (AECs). The AEC phenotype was also confirmed by specific binding of marker lectins (Maclura pomifera and Helix pomatia) and by studying gene expression and activity of cytochrome P450 monooxygenases. Testosterone, ethoxyresorufin, benzyloxyresorufin and verapamil were used as substrates for cytochrome P450-catalysed oxidations and cultured cells were found to be differentiated as well as metabolically competent during cultivation. Therefore, this culture system enables in depth pulmonary biotransformation and toxicity studies.
Collapse
Affiliation(s)
- M Blickwede
- Department of Drug Research and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | | |
Collapse
|