1
|
Shen X, Li M, Wang C, Liu Z, Wu K, Wang A, Bi C, Lu S, Long H, Zhu G. Hypoxia is fine-tuned by Hif-1α and regulates mesendoderm differentiation through the Wnt/β-Catenin pathway. BMC Biol 2022; 20:219. [PMID: 36199093 PMCID: PMC9536055 DOI: 10.1186/s12915-022-01423-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/28/2022] [Indexed: 11/10/2022] Open
Abstract
Background Hypoxia naturally happens in embryogenesis and thus serves as an important environmental factor affecting embryo development. Hif-1α, an essential hypoxia response factor, was mostly considered to mediate or synergistically regulate the effect of hypoxia on stem cells. However, the function and relationship of hypoxia and Hif-1α in regulating mesendoderm differentiation remains controversial. Results We here discovered that hypoxia dramatically suppressed the mesendoderm differentiation and promoted the ectoderm differentiation of mouse embryonic stem cells (mESCs). However, hypoxia treatment after mesendoderm was established promoted the downstream differentiation of mesendoderm-derived lineages. These effects of hypoxia were mediated by the repression of the Wnt/β-Catenin pathway and the Wnt/β-Catenin pathway was at least partially regulated by the Akt/Gsk3β axis. Blocking the Wnt/β-Catenin pathway under normoxia using IWP2 mimicked the effects of hypoxia while activating the Wnt/β-Catenin pathway with CHIR99021 fully rescued the mesendoderm differentiation suppression caused by hypoxia. Unexpectedly, Hif-1α overexpression, in contrast to hypoxia, promoted mesendoderm differentiation and suppressed ectoderm differentiation. Knockdown of Hif-1α under normoxia and hypoxia both inhibited the mesendoderm differentiation. Moreover, hypoxia even suppressed the mesendoderm differentiation of Hif-1α knockdown mESCs, further implying that the effects of hypoxia on the mesendoderm differentiation were Hif-1α independent. Consistently, the Wnt/β-Catenin pathway was enhanced by Hif-1α overexpression and inhibited by Hif-1α knockdown. As shown by RNA-seq, unlike hypoxia, the effect of Hif-1α was relatively mild and selectively regulated part of hypoxia response genes, which fine-tuned the effect of hypoxia on mESC differentiation. Conclusions This study revealed that hypoxia is fine-tuned by Hif-1α and regulates the mesendoderm and ectoderm differentiation by manipulating the Wnt/β-Catenin pathway, which contributed to the understanding of hypoxia-mediated regulation of development. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01423-y.
Collapse
Affiliation(s)
- Xiaopeng Shen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China. .,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China. .,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.
| | - Meng Li
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Chunguang Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Zhongxian Liu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Kun Wu
- Institute of Evolution and Marine Biodiversity, KLMME, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Ao Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Chao Bi
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Shan Lu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| | - Hongan Long
- Institute of Evolution and Marine Biodiversity, KLMME, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Anhui Provincial Key Laboratory of the Conservation and Exploitation of Biological Resources, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China.,Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, College of Life Sciences, Anhui Normal University, Wuhu, 241000, Anhui, China
| |
Collapse
|
2
|
Li H, Xu W, Xiang S, Tao L, Fu W, Liu J, Liu W, Xiao Y, Peng L. Defining the Pluripotent Marker Genes for Identification of Teleost Fish Cell Pluripotency During Reprogramming. Front Genet 2022; 13:819682. [PMID: 35222539 PMCID: PMC8874021 DOI: 10.3389/fgene.2022.819682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Pluripotency is a transient state in early embryos, which is regulated by an interconnected network of pluripotency-related genes. The pluripotent state itself seems to be highly dynamic, which leads to significant differences in the description of induced pluripotent stem cells from different species at the molecular level. With the application of cell reprogramming technology in fish, the establishment of a set of molecular standards for defining pluripotency will be important for the research and potential application of induced pluripotent stem cells in fish. In this study, by BLAST search and expression pattern analysis, we screen out four pluripotent genes (Oct4, Nanog, Tdgf1, and Gdf3) in zebrafish (Danio rerio) and crucian carp (Carassius). These genes were highly expressed in the short period of early embryonic development, but significantly down-regulated after differentiation. Moreover, three genes (Oct4, Nanog and Tdgf1) have been verified that are suitable for identifying the pluripotency of induced pluripotent stem cells in zebrafish and crucian carp. Our study expands the understanding of the pluripotent markers of induced pluripotent stem cells in fish.
Collapse
Affiliation(s)
- Huajin Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Wenting Xu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Sijia Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Leiting Tao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Wen Fu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Jinhui Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Wenbin Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Yamei Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
| | - Liangyue Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- School of Life Sciences, Hunan Normal University, Changsha, China
- *Correspondence: Liangyue Peng,
| |
Collapse
|
3
|
Babaei-Abraki S, Karamali F, Nasr-Esfahani MH. The Role of Endoplasmic Reticulum and Mitochondria in Maintaining Redox Status and Glycolytic Metabolism in Pluripotent Stem Cells. Stem Cell Rev Rep 2022; 18:1789-1808. [PMID: 35141862 DOI: 10.1007/s12015-022-10338-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
Abstract
Pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells (iPSCs), can be applicable for regenerative medicine. They strangely rely on glycolysis metabolism akin to aerobic glycolysis in cancer cells. Upon differentiation, PSCs undergo a metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS). The metabolic shift depends on organelles maturation, transcriptome modification, and metabolic switching. Besides, metabolism-driven chromatin regulation is necessary for cell survival, self-renewal, proliferation, senescence, and differentiation. In this respect, mitochondria may serve as key organelle to adapt environmental changes with metabolic intermediates which are necessary for maintaining PSCs identity. The endoplasmic reticulum (ER) is another organelle whose role in cellular identity remains under-explored. The purpose of our article is to highlight the recent progress on these two organelles' role in maintaining PSCs redox status focusing on metabolism. Topics include redox status, metabolism regulation, mitochondrial dynamics, and ER stress in PSCs. They relate to the maintenance of stem cell properties and subsequent differentiation of stem cells into specific cell types.
Collapse
Affiliation(s)
- Shahnaz Babaei-Abraki
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.,Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
4
|
Karkampouna S, van der Helm D, Scarpa M, van Hoek B, Verspaget HW, Goumans MJ, Coenraad MJ, Kruithof BP, Kruithof-de Julio M. Oncofetal Protein CRIPTO Is Involved in Wound Healing and Fibrogenesis in the Regenerating Liver and Is Associated with the Initial Stages of Cardiac Fibrosis. Cells 2021; 10:3325. [PMID: 34943832 PMCID: PMC8699799 DOI: 10.3390/cells10123325] [Citation(s) in RCA: 3] [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: 10/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022] Open
Abstract
Oncofetal protein, CRIPTO, is silenced during homeostatic postnatal life and often re-expressed in different neoplastic processes, such as hepatocellular carcinoma. Given the reactivation of CRIPTO in pathological conditions reported in various adult tissues, the aim of this study was to explore whether CRIPTO is expressed during liver fibrogenesis and whether this is related to the disease severity and pathogenesis of fibrogenesis. Furthermore, we aimed to identify the impact of CRIPTO expression on fibrogenesis in organs with high versus low regenerative capacity, represented by murine liver fibrogenesis and adult murine heart fibrogenesis. Circulating CRIPTO levels were measured in plasma samples of patients with cirrhosis registered at the waitlist for liver transplantation (LT) and 1 year after LT. The expression of CRIPTO and fibrotic markers (αSMA, collagen type I) was determined in human liver tissues of patients with cirrhosis (on a basis of viral hepatitis or alcoholic disease), in cardiac tissue samples of patients with end-stage heart failure, and in mice with experimental liver and heart fibrosis using immuno-histochemical stainings and qPCR. Mouse models with experimental chronic liver fibrosis, induced with multiple shots of carbon tetrachloride (CCl4) and acute liver fibrosis (one shot of CCl4), were evaluated for CRIPTO expression and fibrotic markers. CRIPTO was overexpressed in vivo (Adenoviral delivery) or functionally sequestered by ALK4Fc ligand trap in the acute liver fibrosis mouse model. Murine heart tissues were evaluated for CRIPTO and fibrotic markers in three models of heart injury following myocardial infarction, pressure overload, and ex vivo induced fibrosis. Patients with end-stage liver cirrhosis showed elevated CRIPTO levels in plasma, which decreased 1 year after LT. Cripto expression was observed in fibrotic tissues of patients with end-stage liver cirrhosis and in patients with heart failure. The expression of CRIPTO in the liver was found specifically in the hepatocytes and was positively correlated with the Model for End-stage Liver Disease (MELD) score for end-stage liver disease. CRIPTO expression in the samples of cardiac fibrosis was limited and mostly observed in the interstitial cells. In the chronic and acute mouse models of liver fibrosis, CRIPTO-positive cells were observed in damaged liver areas around the central vein, which preceded the expression of αSMA-positive stellate cells, i.e., mediators of fibrosis. In the chronic mouse models, the fibrosis and CRIPTO expression were still present after 11 weeks, whereas in the acute model the liver regenerated and the fibrosis and CRIPTO expression resolved. In vivo overexpression of CRIPTO in this model led to an increase in fibrotic markers, while blockage of CRIPTO secreted function inhibited the extent of fibrotic areas and marker expression (αSMA, Collagen type I and III) and induced higher proliferation of residual healthy hepatocytes. CRIPTO expression was also upregulated in several mouse models of cardiac fibrosis. During myocardial infarction CRIPTO is upregulated initially in cardiac interstitial cells, followed by expression in αSMA-positive myofibroblasts throughout the infarct area. After the scar formation, CRIPTO expression decreased concomitantly with the αSMA expression. Temporal expression of CRIPTO in αSMA-positive myofibroblasts was also observed surrounding the coronary arteries in the pressure overload model of cardiac fibrosis. Furthermore, CRIPTO expression was upregulated in interstitial myofibroblasts in hearts cultured in an ex vivo model for cardiac fibrosis. Our results are indicative for a functional role of CRIPTO in the induction of fibrogenesis as well as a potential target in the antifibrotic treatments and stimulation of tissue regeneration.
Collapse
Affiliation(s)
- Sofia Karkampouna
- Department for Biomedical Research, Urology Research Laboratory, Bern University, 3008 Bern, Switzerland; (S.K.); (M.S.)
| | - Danny van der Helm
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.v.d.H.); (B.v.H.); (H.W.V.); (M.J.C.)
| | - Mario Scarpa
- Department for Biomedical Research, Urology Research Laboratory, Bern University, 3008 Bern, Switzerland; (S.K.); (M.S.)
| | - Bart van Hoek
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.v.d.H.); (B.v.H.); (H.W.V.); (M.J.C.)
| | - Hein W. Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.v.d.H.); (B.v.H.); (H.W.V.); (M.J.C.)
| | - Marie-Jose Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (M.-J.G.); (B.P.T.K.)
| | - Minneke J. Coenraad
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (D.v.d.H.); (B.v.H.); (H.W.V.); (M.J.C.)
| | - Boudewijn P.T. Kruithof
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; (M.-J.G.); (B.P.T.K.)
- Department of Cardiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marianna Kruithof-de Julio
- Department for Biomedical Research, Urology Research Laboratory, Bern University, 3008 Bern, Switzerland; (S.K.); (M.S.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Translational Organoid Resource Core, Department for BioMedical Research, Bern University, 3008 Bern, Switzerland
- Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, 3010 Bern, Switzerland
| |
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: 2] [Impact Index Per Article: 0.7] [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
|
Response of Pluripotent Stem Cells to Environmental Stress and Its Application for Directed Differentiation. BIOLOGY 2021; 10:biology10020084. [PMID: 33498611 PMCID: PMC7912122 DOI: 10.3390/biology10020084] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Simple Summary Environmental changes in oxygen concentration, temperature, and mechanical stimulation lead to the activation of specific transcriptional factors and induce the expression of each downstream gene. In general, these responses are protective machinery against such environmental stresses, while these transcriptional factors also regulate cell proliferation, differentiation, and organ development in mammals. In the case of pluripotent stem cells, similar response mechanisms normally work and sometimes stimulate the differentiation cues. Up to now, differentiation protocols utilizing such environmental stresses have been reported to obtain various types of somatic cells from pluripotent stem cells. Basically, environmental stresses as hypoxia (low oxygen), hyperoxia, (high oxygen) and mechanical stress from cell culture plates are relatively safer than chemicals and gene transfers, which affect the genome irreversibly. Therefore, protocols designed with such environments in mind could be useful for the technology development of cell therapy and regenerative medicine. In this manuscript, we summarize recent findings of environmental stress-induced differentiation protocols and discuss their mechanisms. Abstract Pluripotent stem cells have unique characteristics compared to somatic cells. In this review, we summarize the response to environmental stresses (hypoxic, oxidative, thermal, and mechanical stresses) in embryonic stem cells (ESCs) and their applications in the differentiation methods directed to specific lineages. Those stresses lead to activation of each specific transcription factor followed by the induction of downstream genes, and one of them regulates lineage specification. In short, hypoxic stress promotes the differentiation of ESCs to mesodermal lineages via HIF-1α activation. Concerning mechanical stress, high stiffness tends to promote mesodermal differentiation, while low stiffness promotes ectodermal differentiation via the modulation of YAP1. Furthermore, each step in the same lineage differentiation favors each appropriate stiffness of culture plate; for example, definitive endoderm favors high stiffness, while pancreatic progenitor favors low stiffness during pancreatic differentiation of human ESCs. Overall, treatments utilizing those stresses have no genotoxic or carcinogenic effects except oxidative stress; therefore, the differentiated cells are safe and could be useful for cell replacement therapy. In particular, the effect of mechanical stress on differentiation is becoming attractive for the field of regenerative medicine. Therefore, the development of a stress-mediated differentiation protocol is an important matter for the future.
Collapse
|
7
|
Mennen RH, de Leeuw VC, Piersma AH. Oxygen tension influences embryonic stem cell maintenance and has lineage specific effects on neural and cardiac differentiation. Differentiation 2020; 115:1-10. [PMID: 32738735 DOI: 10.1016/j.diff.2020.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
The importance of oxygen tension in in vitro cultures and its effect on embryonic stem cell (ESC) differentiation has been widely acknowledged. Research has mainly focussed on ESC maintenance or on one line of differentiation and only few studies have examined the potential relation between oxygen tension during ESC maintenance and differentiation. In this study we investigated the influence of atmospheric (20%) versus physiologic (5%) oxygen tension in ESC cultures and their differentiation within the cardiac and neural embryonic stem cell tests (ESTc, ESTn). Oxygen tension was set at 5% or 20% and cells were kept in these conditions from starting up cell culture until use for differentiation. Under these oxygen tensions, ESC culture showed no differences in proliferation and gene and protein expression levels. Differentiation was either performed in the same or in the alternative oxygen tension compared to ESC culture creating four different experimental conditions. Cardiac differentiation in 5% instead of 20% oxygen resulted in reduced development of spontaneously beating cardiomyocytes and lower expression of cardiac markers Nkx2.5, Myh6 and MF20 (myosin), regardless whether ESC had been cultured in 5% or 20% oxygen tension. As compared to the control (20% oxygen during stem cell maintenance and differentiation), neural differentiation in 5% oxygen with ESC cultured in 20% oxygen led to more cardiac and neural crest cell differentiation. The opposite experimental condition of neural differentiation in 20% oxygen with ESC cultured in 5% oxygen resulted in more glial differentiation. ESC that were maintained and differentiated in 5% oxygen showed an increase in neural crest and oligodendrocytes as compared to 20% oxygen during stem cell maintenance and differentiation. This study showed major effects on ESC differentiation in ESTc and ESTn of oxygen tension, which is an important variable to consider when designing and developing a stem cell-based in vitro system.
Collapse
Affiliation(s)
- Regina H Mennen
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - Victoria C de Leeuw
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
8
|
Rodrigues Sousa E, Zoni E, Karkampouna S, La Manna F, Gray PC, De Menna M, Kruithof-de Julio M. A Multidisciplinary Review of the Roles of Cripto in the Scientific Literature Through a Bibliometric Analysis of its Biological Roles. Cancers (Basel) 2020; 12:cancers12061480. [PMID: 32517087 PMCID: PMC7352664 DOI: 10.3390/cancers12061480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Cripto is a small glycosylphosphatidylinisitol (GPI)-anchored and secreted oncofetal protein that plays important roles in regulating normal physiological processes, including stem cell differentiation, embryonal development, and tissue growth and remodeling, as well as pathological processes such as tumor initiation and progression. Cripto functions as a co-receptor for TGF-β ligands such as Nodal, GDF1, and GDF3. Soluble and secreted forms of Cripto also exhibit growth factor-like activity and activate SRC/MAPK/PI3K/AKT pathways. Glucose-Regulated Protein 78 kDa (GRP78) binds Cripto at the cell surface and has been shown to be required for Cripto signaling via both TGF-β and SRC/MAPK/PI3K/AKT pathways. To provide a comprehensive overview of the scientific literature related to Cripto, we performed, for the first time, a bibliometric analysis of the biological roles of Cripto as reported in the scientific literature covering the last 10 years. We present different fields of knowledge in comprehensive areas of research on Cripto, ranging from basic to translational research, using a keyword-driven approach. Our ultimate aim is to aid the scientific community in conducting targeted research by identifying areas where research has been conducted so far and, perhaps more importantly, where critical knowledge is still missing.
Collapse
Affiliation(s)
- Elisa Rodrigues Sousa
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Eugenio Zoni
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Sofia Karkampouna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Federico La Manna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Marta De Menna
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
| | - Marianna Kruithof-de Julio
- Department for Biomedical Research, Urology Research Laboratory, University of Bern, 3008 Bern, Switzerland; (E.R.S.); (E.Z.); (S.K.); (F.L.M.); (M.D.M.)
- Department of Urology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
9
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
10
|
Terry S, Faouzi Zaarour R, Hassan Venkatesh G, Francis A, El-Sayed W, Buart S, Bravo P, Thiery J, Chouaib S. Role of Hypoxic Stress in Regulating Tumor Immunogenicity, Resistance and Plasticity. Int J Mol Sci 2018; 19:ijms19103044. [PMID: 30301213 PMCID: PMC6213127 DOI: 10.3390/ijms19103044] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/28/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022] Open
Abstract
Hypoxia, or gradients of hypoxia, occurs in most growing solid tumors and may result in pleotropic effects contributing significantly to tumor aggressiveness and therapy resistance. Indeed, the generated hypoxic stress has a strong impact on tumor cell biology. For example, it may contribute to increasing tumor heterogeneity, help cells gain new functional properties and/or select certain cell subpopulations, facilitating the emergence of therapeutic resistant cancer clones, including cancer stem cells coincident with tumor relapse and progression. It controls tumor immunogenicity, immune plasticity, and promotes the differentiation and expansion of immune-suppressive stromal cells. In this context, manipulation of the hypoxic microenvironment may be considered for preventing or reverting the malignant transformation. Here, we review the current knowledge on how hypoxic stress in tumor microenvironments impacts on tumor heterogeneity, plasticity and resistance, with a special interest in the impact on immune resistance and tumor immunogenicity.
Collapse
Affiliation(s)
- Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine-Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| | - Rania Faouzi Zaarour
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Amirtharaj Francis
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Walid El-Sayed
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| | - Stéphanie Buart
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine-Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| | - Pamela Bravo
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine-Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| | - Jérome Thiery
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine-Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine-Univ. Paris-Sud, University Paris-Saclay, Villejuif F-94805, France.
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates.
| |
Collapse
|
11
|
Kim S, Yoon YM, Han YS, Lee JH, Hur J, Lee SH. Administration of Cripto in GRP78 overexpressed human MSCs enhances stem cell viability and angiogenesis during human MSC transplantation therapy. Cell Prolif 2018; 51:e12463. [PMID: 29722092 DOI: 10.1111/cpr.12463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/24/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The purpose of this study was to explore the effectiveness of concurrent GRP78 overexpression combined with Cripto on hMSC proliferation and migration both in vitro and in vivo. Specifically, we explored whether the treatment enhances effectiveness of hMSC transplantation in ischaemic tissue. MATERIALS AND METHODS Human MSCs obtained from human adipose tissue were cultured in α-minimum essential medium (Hyclone, Logan, UT, USA) supplemented with 10% (v/v) foetal bovine serum (Hyclone), 100 U mL-1 penicillin and 100 μg mL-1 streptomycin. Murine hindlimb ischaemic model was generated with 8-week-old male nude BALB/c mice (Biogenomics, Seoul, Korea) maintained under a 12-h light/dark cycle following the established protocol with minor modification. Cellular injection was performed no later than 3 hour after surgery. Lipofectamine transfection, single-cell cultivation assay, transwell assay, scratch wound-healing migration assay, immunohistochemistry and western blotting assays were performed. RESULTS Overexpression of GRP78 along with Cripto enhanced hMSC proliferation, migration and invasion. It increased interaction of surface GRP78 receptor with Cripto via JAK2/STAT3 pathway. We confirmed our proposed mechanism by showing that treatment with GRP78 antibody blocks the enhancement in vitro. In vivo, we observed that Cripto induced by the hypoxic environment in hindlimb ischaemic model interacts with the overexpressed GRP78 and increases hMSC proliferation, migration and invasion potentials as well as angiogenesis around transplanted ischaemic site via cytokine secretions. CONCLUSIONS These results demonstrate supporting evidences that GRP78-Cripto combination technique offers novel strategy to enhance MSC proliferation, migration and invasion potentials as well as angiogenesis around ischaemic site, ultimately facilitating MSC-based transplantation therapy in ischaemic conditions.
Collapse
Affiliation(s)
- S Kim
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Y M Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Y-S Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - J H Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - J Hur
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea
| | - S H Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
12
|
Park SW, Do HJ, Han MH, Choi W, Kim JH. The expression of the embryonic gene Cripto-1 is regulated by OCT4 in human embryonal carcinoma NCCIT cells. FEBS Lett 2017; 592:24-35. [PMID: 29223130 DOI: 10.1002/1873-3468.12935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 01/23/2023]
Abstract
Cripto-1 and OCT4, expressed in stem cells and cancers, play important roles in tumorigenesis. Here, we demonstrate that Cripto-1 expression is regulated by OCT4 in human embryonic carcinoma NCCIT cells. The endogenous expression of Cripto-1 and OCT4 is significantly reduced during differentiation. Cripto-1 expression is increased by OCT4 overexpression, but decreased by shRNA-mediated OCT4 knockdown. OCT4 overexpression significantly activates Cripto-1 transcriptional activity. A 5'-upstream minimal promoter sequence in the gene-encoding Cripto-1 is significantly activated by OCT4 overexpression. Mutation of the putative OCT4-binding site abolishes OCT4-mediated activation of the Cripto-1 promoter. The OCT4 transactivation domains mediate transcriptional activity of the Cripto-1 minimal promoter through direct interaction. Taken together, OCT4 plays an important role as a transcriptional activator of Cripto-1 expression in NCCIT cells.
Collapse
Affiliation(s)
- Sung-Won Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-Si, Gyeonggi-Do, Korea
| | - Hyun-Jin Do
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-Si, Gyeonggi-Do, Korea
| | - Mi-Hee Han
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-Si, Gyeonggi-Do, Korea
| | - Wonbin Choi
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-Si, Gyeonggi-Do, Korea
| | - Jae-Hwan Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-Si, Gyeonggi-Do, Korea
| |
Collapse
|
13
|
Talkhabi M, Zonooz ER, Baharvand H. Boosters and barriers for direct cardiac reprogramming. Life Sci 2017; 178:70-86. [PMID: 28427897 DOI: 10.1016/j.lfs.2017.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/08/2017] [Accepted: 04/16/2017] [Indexed: 12/16/2022]
Abstract
Heart disease is currently the most significant cause of morbidity and mortality worldwide, which accounts for approximately 33% of all deaths. Recently, a promising and alchemy-like strategy has been developed called direct cardiac reprogramming, which directly converts somatic cells such as fibroblasts to cardiac lineage cells such as cardiomyocytes (CMs), termed induced CMs or iCMs. The first in vitro cardiac reprogramming study, mediated by cardiac transcription factors (TFs)-Gata4, Tbx5 and Mef2C-, was not enough efficient to produce an adequate number of fully reprogrammed, functional iCMs. As a result, numerous combinations of cardiac TFs exist for direct cardiac reprogramming of mouse and human fibroblasts. However, the efficiency of direct cardiac reprogramming remains low. Recently, a number of cellular and molecular mechanisms have been identified to increase the efficiency of direct cardiac reprogramming and the quality of iCMs. For example, microgrooved substrate, cardiogenic growth factors [VEGF, FGF, BMP4 and Activin A], and an appropriate stoichiometry of TFs boost the direct cardiac reprogramming. On the other hand, serum, TGFβ signaling, activators of epithelial to mesenchymal transition, and some epigenetic factors (Bmi1 and Ezh2) are barriers for direct cardiac reprogramming. Manipulating these mechanisms by the application of boosters and removing barriers can increase the efficiency of direct cardiac reprogramming and possibly make iCMs reliable for cell-based therapy or other potential applications. In this review, we summarize the latest trends in cardiac TF- or miRNA-based direct cardiac reprogramming and comprehensively discuses all molecular and cellular boosters and barriers affecting direct cardiac reprogramming.
Collapse
Affiliation(s)
- Mahmood Talkhabi
- Department of Animal Sciences and Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Elmira Rezaei Zonooz
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
14
|
Kirabo A, Ryzhov S, Gupte M, Sengsayadeth S, Gumina RJ, Sawyer DB, Galindo CL. Neuregulin-1β induces proliferation, survival and paracrine signaling in normal human cardiac ventricular fibroblasts. J Mol Cell Cardiol 2017; 105:59-69. [PMID: 28263756 PMCID: PMC5715731 DOI: 10.1016/j.yjmcc.2017.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/14/2017] [Accepted: 03/01/2017] [Indexed: 01/27/2023]
Abstract
Neuregulin-1β (NRG-1β) is critical for cardiac development and repair, and recombinant forms are currently being assessed as possible therapeutics for systolic heart failure. We previously demonstrated that recombinant NRG-1β reduces cardiac fibrosis in an animal model of cardiac remodeling and heart failure, suggesting that there may be direct effects on cardiac fibroblasts. Here we show that NRG-1β receptors (ErbB2, ErbB3, and ErbB4) are expressed in normal human cardiac ventricular (NHCV) fibroblast cell lines. Treatment of NHCV fibroblasts with recombinant NRG-1β induced activation of the AKT pathway, which was phosphoinositide 3-kinase (PI3K)-dependent. Moreover, the NRG-1β-induced PI3K/AKT signaling in these cells required phosphorylation of both ErbB2 and ErbB3 receptors at tyrosine (Tyr)1248 and Tyr1289 respectively. RNASeq analysis of NRG-1β-treated cardiac fibroblasts obtained from three different individuals revealed a global gene expression signature consistent with cell growth and survival. We confirmed enhanced cellular proliferation and viability in NHCV fibroblasts in response to NRG-1β, which was abrogated by PI3K, ErbB2, and ErbB3 inhibitors. NRG-1β also induced production and secretion of cytokines (interleukin-1α and interferon-γ) and pro-reparative factors (angiopoietin-2, brain-derived neurotrophic factor, and crypto-1), suggesting a role in cardiac repair through the activation of paracrine signaling.
Collapse
Affiliation(s)
- Annet Kirabo
- Department of Pharmacology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Sergey Ryzhov
- Maine Medical Research Institute, 81 Research Drive, Scarborough, ME 04074, United States.
| | - Manisha Gupte
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Seng Sengsayadeth
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States; Department of Pathology, Immunology, and Microbiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| | - Douglas B Sawyer
- Maine Medical Research Institute, 81 Research Drive, Scarborough, ME 04074, United States.
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States.
| |
Collapse
|
15
|
Activin A Modulates CRIPTO-1/HNF4 α+ Cells to Guide Cardiac Differentiation from Human Embryonic Stem Cells. Stem Cells Int 2017; 2017:4651238. [PMID: 28163723 PMCID: PMC5253508 DOI: 10.1155/2017/4651238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/29/2016] [Accepted: 12/01/2016] [Indexed: 01/02/2023] Open
Abstract
The use of human pluripotent stem cells in basic and translational cardiac research requires efficient differentiation protocols towards cardiomyocytes. In vitro differentiation yields heterogeneous populations of ventricular-, atrial-, and nodal-like cells hindering their potential applications in regenerative therapies. We described the effect of the growth factor Activin A during early human embryonic stem cell fate determination in cardiac differentiation. Addition of high levels of Activin A during embryoid body cardiac differentiation augmented the generation of endoderm derivatives, which in turn promoted cardiomyocyte differentiation. Moreover, a dose-dependent increase in the coreceptor expression of the TGF-β superfamily member CRIPTO-1 was observed in response to Activin A. We hypothesized that interactions between cells derived from meso- and endodermal lineages in embryoid bodies contributed to improved cell maturation in early stages of cardiac differentiation, improving the beating frequency and the percentage of contracting embryoid bodies. Activin A did not seem to affect the properties of cardiomyocytes at later stages of differentiation, measuring action potentials, and intracellular Ca2+ dynamics. These findings are relevant for improving our understanding on human heart development, and the proposed protocol could be further explored to obtain cardiomyocytes with functional phenotypes, similar to those observed in adult cardiac myocytes.
Collapse
|
16
|
Ngo D, Sinha S, Shen D, Kuhn EW, Keyes MJ, Shi X, Benson MD, O'Sullivan JF, Keshishian H, Farrell LA, Fifer MA, Vasan RS, Sabatine MS, Larson MG, Carr SA, Wang TJ, Gerszten RE. Aptamer-Based Proteomic Profiling Reveals Novel Candidate Biomarkers and Pathways in Cardiovascular Disease. Circulation 2016; 134:270-85. [PMID: 27444932 DOI: 10.1161/circulationaha.116.021803] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/20/2016] [Indexed: 01/22/2023]
Abstract
BACKGROUND Single-stranded DNA aptamers are oligonucleotides of ≈50 base pairs in length selected for their ability to bind proteins with high specificity and affinity. Emerging DNA aptamer-based technologies may address limitations of existing proteomic techniques, including low sample throughput, which have hindered proteomic analyses of large cohorts. METHODS To identify early biomarkers of myocardial injury, we applied an aptamer-based proteomic platform that measures 1129 proteins to a clinically relevant perturbational model of planned myocardial infarction (PMI), patients undergoing septal ablation for hypertrophic cardiomyopathy. Blood samples were obtained before and at 10 and 60 minutes after PMI, and protein changes were assessed by repeated-measures analysis of variance. The generalizability of our PMI findings was evaluated in a spontaneous myocardial infarction cohort (Wilcoxon rank-sum). We then tested the platform's ability to detect associations between proteins and Framingham Risk Score components in the Framingham Heart Study, performing regression analyses for each protein versus each clinical trait. RESULTS We found 217 proteins that significantly changed in the peripheral vein blood after PMI in a derivation cohort (n=15; P<5.70E-5). Seventy-nine of these proteins were validated in an independent PMI cohort (n=15; P<2.30E-4); >85% were directionally consistent and reached nominal significance. We detected many protein changes that are novel in the context of myocardial injury, including Dickkopf-related protein 4, a WNT pathway inhibitor (peak increase 124%, P=1.29E-15) and cripto, a growth factor important in cardiac development (peak increase 64%, P=1.74E-4). Among the 40 validated proteins that increased within 1 hour after PMI, 23 were also elevated in patients with spontaneous myocardial infarction (n=46; P<0.05). Framingham Heart Study analyses revealed 156 significant protein associations with the Framingham Risk Score (n=899), including aminoacylase 1 (β=0.3386, P=2.54E-22) and trigger factor 2 (β=0.2846, P=5.71E-17). Furthermore, we developed a novel workflow integrating DNA-based immunoaffinity with mass spectrometry to analytically validate aptamer specificity. CONCLUSIONS Our results highlight an emerging proteomics tool capable of profiling >1000 low-abundance analytes with high sensitivity and high precision, applicable both to well-phenotyped perturbational studies and large human cohorts, as well.
Collapse
Affiliation(s)
- Debby Ngo
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Sumita Sinha
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Dongxiao Shen
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Eric W Kuhn
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Michelle J Keyes
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Xu Shi
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Mark D Benson
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - John F O'Sullivan
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Hasmik Keshishian
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Laurie A Farrell
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Michael A Fifer
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Ramachandran S Vasan
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Marc S Sabatine
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Martin G Larson
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| | - Steven A Carr
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.).
| | - Thomas J Wang
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.).
| | - Robert E Gerszten
- From Division of Pulmonary and Critical Care Medicine, Department of Medicine (D.N.) and the Cardiovascular Research Center (D.N., S.S., D.S., M.J.K., X.S., M.D.B., J.F.O., L.A.F., R.E.G.), Massachusetts General Hospital, Harvard Medical School, Boston; Broad Institute of MIT and Harvard, Cambridge, MA (E.W.K., H.K., S.A.C., R.E.G.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.D.B., M.S.S.); Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston (M.A.F., M.S.S., R.E.G.); Preventive Medicine Section, Department of Medicine, Boston University School of Medicine, MA (R.S.V.); Department of Biostatistics, Boston University School of Public Health, MA (M.G.L.); The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA (M.G.L.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (R.E.G.)
| |
Collapse
|
17
|
Ectopic expression of Cripto-1 in transgenic mouse embryos causes hemorrhages, fatal cardiac defects and embryonic lethality. Sci Rep 2016; 6:34501. [PMID: 27687577 PMCID: PMC5043281 DOI: 10.1038/srep34501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/15/2016] [Indexed: 12/23/2022] Open
Abstract
Targeted disruption of Cripto-1 in mice caused embryonic lethality at E7.5, whereas we unexpectedly found that ectopic Cripto-1 expression in mouse embryos also led to embryonic lethality, which prompted us to characterize the causes and mechanisms underlying embryonic death due to ectopic Cripto-1 expression. RCLG/EIIa-Cre embryos displayed complex phenotypes between embryonic day 14.5 (E14.5) and E17.5, including fatal hemorrhages (E14.5-E15.5), embryo resorption (E14.5-E17.5), pale body surface (E14.5-E16.5) and no abnormal appearance (E14.5-E16.5). Macroscopic and histological examination revealed that ectopic expression of Cripto-1 transgene in RCLG/EIIa-Cre embryos resulted in lethal cardiac defects, as evidenced by cardiac malformations, myocardial thinning, failed assembly of striated myofibrils and lack of heartbeat. In addition, Cripto-1 transgene activation beginning after E8.5 also caused the aforementioned lethal cardiac defects in mouse embryos. Furthermore, ectopic Cripto-1 expression in embryonic hearts reduced the expression of cardiac transcription factors, which is at least partially responsible for the aforementioned lethal cardiac defects. Our results suggest that hemorrhages and cardiac abnormalities are two important lethal factors in Cripto-1 transgenic mice. Taken together, these findings are the first to demonstrate that sustained Cripto-1 transgene expression after E11.5 causes fatal hemorrhages and lethal cardiac defects, leading to embryonic death at E14.5-17.5.
Collapse
|
18
|
HIF-1alpha Deficiency Attenuates the Cardiomyogenesis of Mouse Embryonic Stem Cells. PLoS One 2016; 11:e0158358. [PMID: 27355368 PMCID: PMC4927095 DOI: 10.1371/journal.pone.0158358] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/14/2016] [Indexed: 01/09/2023] Open
Abstract
Cardiac cell formation, cardiomyogenesis, is critically dependent on oxygen availability. It is known that hypoxia, a reduced oxygen level, modulates the in vitro differentiation of pluripotent cells into cardiomyocytes via hypoxia inducible factor-1alpha (HIF-1α)-dependent mechanisms. However, the direct impact of HIF-1α deficiency on the formation and maturation of cardiac-like cells derived from mouse embryonic stem cells (mESC) in vitro remains to be elucidated. In the present study, we demonstrated that HIF-1α deficiency significantly altered the quality and quantity of mESC-derived cardiomyocytes. It was accompanied with lower mRNA and protein levels of cardiac cell specific markers (myosin heavy chains 6 and 7) and with a decreasing percentage of myosin heavy chain α and β, and cardiac troponin T-positive cells. As to structural aspects of the differentiated cardiomyocytes, the localization of contractile proteins (cardiac troponin T, myosin heavy chain α and β) and the organization of myofibrils were also different. Simultaneously, HIF-1α deficiency was associated with a lower percentage of beating embryoid bodies. Interestingly, an observed alteration in the in vitro differentiation scheme of HIF-1α deficient cells was accompanied with significantly lower expression of the endodermal marker (hepatic nuclear factor 4 alpha). These findings thus suggest that HIF-1α deficiency attenuates spontaneous cardiomyogenesis through the negative regulation of endoderm development in mESC differentiating in vitro.
Collapse
|
19
|
Oxygen cycling to improve survival of stem cells for myocardial repair: A review. Life Sci 2016; 153:124-31. [PMID: 27091653 DOI: 10.1016/j.lfs.2016.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/28/2016] [Accepted: 04/08/2016] [Indexed: 02/08/2023]
Abstract
Heart disease represents the leading cause of death among Americans. There is currently no clinical treatment to regenerate viable myocardium following myocardial infarction, and patients may suffer progressive deterioration and decreased myocardial function from the effects of remodeling of the necrotic myocardium. New therapeutic strategies hold promise for patients who suffer from ischemic heart disease by directly addressing the restoration of functional myocardium following death of cardiomyocytes. Therapeutic stem cell transplantation has shown modest benefit in clinical human trials with decreased fibrosis and increased functional myocardium. Moreover, autologous transplantation holds the potential to implement these therapies while avoiding the immunomodulation concerns of heart transplantation. Despite these benefits, stem cell therapy has been characterized by poor survival and low engraftment of injected stem cells. The hypoxic tissue environment of the ischemic/infracting myocardium impedes stem cell survival and engraftment in myocardial tissue. Hypoxic preconditioning has been suggested as a viable strategy to increase hypoxic tolerance of stem cells. A number of in vivo and in vitro studies have demonstrated improved stem cell viability by altering stem cell secretion of protein signals and up-regulation of numerous paracrine signaling pathways that affect inflammatory, survival, and angiogenic signaling pathways. This review will discuss both the mechanisms of hypoxic preconditioning as well as the effects of hypoxic preconditioning in different cell and animal models, examining the pitfalls in current research and the next steps into potentially implementing this methodology in clinical research trials.
Collapse
|
20
|
The stabilization of hypoxia inducible factor modulates differentiation status and inhibits the proliferation of mouse embryonic stem cells. Chem Biol Interact 2016; 244:204-14. [DOI: 10.1016/j.cbi.2015.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/26/2015] [Accepted: 12/17/2015] [Indexed: 01/16/2023]
|
21
|
Klauzinska M, Bertolette D, Tippireddy S, Strizzi L, Gray PC, Gonzales M, Duroux M, Ruvo M, Wechselberger C, Castro NP, Rangel MC, Focà A, Sandomenico A, Hendrix MJC, Salomon D, Cuttitta F. Cripto-1: an extracellular protein - connecting the sequestered biological dots. Connect Tissue Res 2015; 56:364-80. [PMID: 26327334 DOI: 10.3109/03008207.2015.1077239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cripto-1 (CR-1) is a multifunctional embryonic protein that is re-expressed during inflammation, wound repair, and malignant transformation. CR-1 can function either as a tethered co-receptor or shed as a free ligand underpinning its flexible role in cell physiology. CR-1 has been shown to mediate cell growth, migration, invasion, and induce epithelial to mesenchymal transition (EMT). The main signaling pathways mediating CR-1 effects include Nodal-dependent (Smad2/3) and Nodal-independent (Src/p44/42/Akt) signaling transduction pathways. In addition, there are several naturally occurring binding partner proteins (BPPs) for CR-1 that can either agonize or antagonize its bioactivity. We will review the collective role of CR-1 as an extracellular protein, discuss caveats to consider in developing a quantitation assay, define possible mechanistic avenues applicable for drug discovery, and report on our experimental approaches to overcome these problematic issues.
Collapse
Affiliation(s)
- Malgorzata Klauzinska
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Daniel Bertolette
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Sudhamsh Tippireddy
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Luigi Strizzi
- b Department of Pathology , Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Peter C Gray
- c Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies , La Jolla , CA , USA
| | - Monica Gonzales
- d Office of Research Operations, Office of the Director, Center for Cancer Research, National Cancer Institute , Bethesda , MD , USA
| | - Meg Duroux
- e Laboratory of Cancer Biology , Biomedicine Group, Department of Health Science and Technology, Aalborg University , Aalborg East , Denmark
| | - Menotti Ruvo
- f CIRPeB, University of Naples Federico II , Napoli , Italy .,g Istituto di Biostrutture e Bioimmagini del CRN , Napoli , Italy
| | | | - Nadia P Castro
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Maria Cristina Rangel
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Annalia Focà
- g Istituto di Biostrutture e Bioimmagini del CRN , Napoli , Italy .,i Dipartimento di Farmacia, University of Naples Federico II , Napoli , Italy , and
| | | | - Mary J C Hendrix
- j Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - David Salomon
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| | - Frank Cuttitta
- a Tumor Growth Factor Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute , Frederick , MD , USA
| |
Collapse
|
22
|
Pilli VS, Gupta K, Kotha BP, Aradhyam GK. Snail-mediated Cripto-1 repression regulates the cell cycle and epithelial-mesenchymal transition-related gene expression. FEBS Lett 2015; 589:1249-56. [PMID: 25889638 DOI: 10.1016/j.febslet.2015.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/23/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
Transcription factor Snail mediates epithelial to mesenchymal transitions (EMT) by coordinate repression of epithelial markers, facilitating mass cell movement during germ layer formation. Aberrant reprogramming in its signaling pathways causes metastatic cancer. Snail's involvement in "fate-changing" decisions is however not understood. Cripto-1 shares a common temporal expression pattern with Snail during development. While Cripto-1 is required for mammary morphogenesis and hematopoietic stem cell renewal, its unregulated expression causes metastatic cancers. Therefore, we suspected that Snail regulates the expression of Cripto-1 controlling decisions such as motility, transformation and differentiation. We demonstrate that Snail represses Cripto-1 gene by direct transcriptional interaction and propose a novel mechanism by which it co-ordinately regulates cell fate decisions during development and could be causal of cancers.
Collapse
Affiliation(s)
- Vijaya S Pilli
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - K Gupta
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - Bhanu P Kotha
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India
| | - Gopala Krishna Aradhyam
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Bioscience, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
23
|
Loying P, Manhas J, Sen S, Bose B. Autoregulation and heterogeneity in expression of human Cripto-1. PLoS One 2015; 10:e0116748. [PMID: 25658584 PMCID: PMC4319928 DOI: 10.1371/journal.pone.0116748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/12/2014] [Indexed: 11/18/2022] Open
Abstract
Cripto-1 (CR-1) is involved in various processes in embryonic development and cancer. Multiple pathways regulate CR-1 expression. Our present work demonstrates a possible positive feedback circuit where CR-1 induces its own expression. Using U-87 MG cells treated with exogenous CR-1, we show that such induction involves ALK4/SMAD2/3 pathway. Stochasticity in gene expression gives rise to heterogeneity in expression in genetically identical cells. Positive feedback increases such heterogeneity and often gives rise to two subpopulations of cells, having higher and lower expression of a gene. Using flow cytometry, we show that U-87 MG cells have a minuscule subpopulation with detectable expression of CR-1. Induction of CR-1 expression, by exogenous CR-1, increases the size of this CR-1 positive subpopulation. However, even at very high dose, most of the cells remain CR-1 negative. We show that population behavior of CR-1 induction has a signature similar to bimodal expression expected in a transcriptional circuit with positive feedback. We further show that treatment of U-87 MG cells with CR-1 leads to higher expression of drug efflux protein MDR-1 in the CR-1 positive subpopulation, indicating correlated induction of these two proteins. Positive feedback driven heterogeneity in expression of CR-1 may play crucial role in phenotypic diversification of cancer cells.
Collapse
Affiliation(s)
- Pojul Loying
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, India
| | - Janvie Manhas
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sudip Sen
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Biplab Bose
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati, India
- * E-mail:
| |
Collapse
|
24
|
Ruggiero D, Nappo S, Nutile T, Sorice R, Talotta F, Giorgio E, Bellenguez C, Leutenegger AL, Liguori GL, Ciullo M. Genetic variants modulating CRIPTO serum levels identified by genome-wide association study in Cilento isolates. PLoS Genet 2015; 11:e1004976. [PMID: 25629528 PMCID: PMC4309561 DOI: 10.1371/journal.pgen.1004976] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/29/2014] [Indexed: 02/07/2023] Open
Abstract
Cripto, the founding member of the EGF-CFC genes, plays an essential role in embryo development and is involved in cancer progression. Cripto is a GPI-anchored protein that can interact with various components of multiple signaling pathways, such as TGF-β, Wnt and MAPK, driving different processes, among them epithelial-mesenchymal transition, cell proliferation, and stem cell renewal. Cripto protein can also be cleaved and released outside the cell in a soluble and still active form. Cripto is not significantly expressed in adult somatic tissues and its re-expression has been observed associated to pathological conditions, mainly cancer. Accordingly, CRIPTO has been detected at very low levels in the plasma of healthy volunteers, whereas its levels are significantly higher in patients with breast, colon or glioblastoma tumors. These data suggest that CRIPTO levels in human plasma or serum may have clinical significance. However, very little is known about the variability of serum levels of CRIPTO at a population level and the genetic contribution underlying this variability remains unknown. Here, we report the first genome-wide association study of CRIPTO serum levels in isolated populations (n = 1,054) from Cilento area in South Italy. The most associated SNPs (p-value<5*10-8) were all located on chromosome 3p22.1-3p21.3, in the CRIPTO gene region. Overall six CRIPTO associated loci were replicated in an independent sample (n = 535). Pathway analysis identified a main network including two other genes, besides CRIPTO, in the associated regions, involved in cell movement and proliferation. The replicated loci explain more than 87% of the CRIPTO variance, with 85% explained by the most associated SNP. Moreover, the functional analysis of the main associated locus identified a causal variant in the 5’UTR of CRIPTO gene which is able to strongly modulate CRIPTO expression through an AP-1-mediate transcriptional regulation. Cripto gene has a fundamental role in embryo development and is also involved in cancer. The protein is bound to the cell membrane through an anchor, that can be cleaved, causing the secretion of the protein, in a still active form. In the adult, CRIPTO is detected at very low levels in normal tissues and in the blood, while its increase in both tissues and blood is associated to pathological conditions, mainly cancer. As other GPI linked proteins such as the carcinoembryonic antigen (CEA), one of the most used tumor markers, CRIPTO is able to reach the bloodstream. Therefore, CRIPTO represents a new promising biomarker and potential therapeutic target, and blood CRIPTO levels might be associated to clinical features. Here we examined the variability of blood CRIPTO levels at a population level (population isolates from the Cilento region in South Italy) and we investigated the genetic architecture underlying this variability. We reported the association of common genetic variants with the levels of CRIPTO protein in the blood and we identified a main locus on chromosome 3 and additional five associated loci. Moreover, through functional analyses, we were able to uncover the mechanism responsible for the variation in CRIPTO levels, which is a regulation mediated by the transcriptional factor AP-1.
Collapse
Affiliation(s)
- Daniela Ruggiero
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Stefania Nappo
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Teresa Nutile
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Francesco Talotta
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Emilia Giorgio
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Celine Bellenguez
- Institut Pasteur de Lille, Lille, France
- Inserm, U744, Lille, France
- Université Lille-Nord de France, Lille, France
| | - Anne-Louise Leutenegger
- Inserm, U946, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, IUH, UMR-S 946, Paris, France
| | - Giovanna L. Liguori
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
| | - Marina Ciullo
- Institute of Genetics and Biophysics A. Buzzati-Traverso, CNR, Naples, Italy
- * E-mail:
| |
Collapse
|
25
|
Zhou Y, Fujisawa I, Ino K, Matsue T, Shiku H. Metabolic suppression during mesodermal differentiation of embryonic stem cells identified by single-cell comprehensive gene expression analysis. MOLECULAR BIOSYSTEMS 2015. [DOI: 10.1039/c5mb00340g] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Metabolic suppression has been revealed during mesodermal differentiation by using single-cell gene expression analysis.
Collapse
Affiliation(s)
- Yuanshu Zhou
- WPI-Advanced Institute for Materials Research
- Tohoku University
- Sendai 980-8577
- Japan
| | - Ikuma Fujisawa
- Graduate School of Environmental Studies
- Tohoku University
- Sendai 980-8579
- Japan
| | - Kosuke Ino
- Graduate School of Environmental Studies
- Tohoku University
- Sendai 980-8579
- Japan
| | - Tomokazu Matsue
- WPI-Advanced Institute for Materials Research
- Tohoku University
- Sendai 980-8577
- Japan
- Graduate School of Environmental Studies
| | - Hitoshi Shiku
- Graduate School of Environmental Studies
- Tohoku University
- Sendai 980-8579
- Japan
| |
Collapse
|
26
|
The multifaceted role of the embryonic gene Cripto-1 in cancer, stem cells and epithelial-mesenchymal transition. Semin Cancer Biol 2014; 29:51-8. [PMID: 25153355 DOI: 10.1016/j.semcancer.2014.08.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/07/2014] [Indexed: 01/04/2023]
Abstract
Cripto-1 (CR-1)/Teratocarcinoma-derived growth factor1 (TDGF-1) is a cell surface glycosylphosphatidylinositol (GPI)-linked glycoprotein that can function either in cis (autocrine) or in trans (paracrine). The cell membrane cis form is found in lipid rafts and endosomes while the trans acting form lacking the GPI anchor is soluble. As a member of the epidermal growth factor (EGF)/Cripto-1-FRL-1-Cryptic (CFC) family, CR-1 functions as an obligatory co-receptor for the transforming growth factor-β (TGF-β) family members, Nodal and growth and differentiation factors 1 and 3 (GDF1/3) by activating Alk4/Alk7 signaling pathways that involve Smads 2, 3 and 4. In addition, CR-1 can activate non-Smad-dependent signaling elements such as PI3K, Akt and MAPK. Both of these pathways depend upon the 78kDa glucose regulated protein (GRP78). Finally, CR-1 can facilitate signaling through the canonical Wnt/β-catenin and Notch/Cbf-1 pathways by functioning as a chaperone protein for LRP5/6 and Notch, respectively. CR-1 is essential for early embryonic development and maintains embryonic stem cell pluripotentiality. CR-1 performs an essential role in the etiology and progression of several types of human tumors where it is expressed in a population of cancer stem cells (CSCs) and facilitates epithelial-mesenchymal transition (EMT). In this context, CR-1 can significantly enhance tumor cell migration, invasion and angiogenesis. Collectively, these facts suggest that CR-1 may be an attractive target in the diagnosis, prognosis and therapy of several types of human cancer.
Collapse
|
27
|
Fynes K, Tostoes R, Ruban L, Weil B, Mason C, Veraitch FS. The differential effects of 2% oxygen preconditioning on the subsequent differentiation of mouse and human pluripotent stem cells. Stem Cells Dev 2014; 23:1910-22. [PMID: 24734982 DOI: 10.1089/scd.2013.0504] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
A major challenge facing the development of effective cell therapies is the efficient differentiation of pluripotent stem cells (PSCs) into pure populations. Lowering oxygen tension to physiological levels can affect both the expansion and differentiation stages. However, to date, there are no studies investigating the knock-on effect of culturing PSCs under low oxygen conditions on subsequent lineage commitment at ambient oxygen levels. PSCs were passaged three times at 2% O2 before allowing cells to spontaneously differentiate as embryoid bodies (EBs) in high oxygen (20% O2) conditions. Maintenance of mouse PSCs in low oxygen was associated with a significant increase in the expression of early differentiation markers FGF5 and Eomes, while conversely we observed decreased expression of these genes in human PSCs. Low oxygen preconditioning primed mouse PSCs for their subsequent differentiation into mesodermal and endodermal lineages, as confirmed by increased gene expression of Eomes, Goosecoid, Brachyury, AFP, Sox17, FoxA2, and protein expression of Brachyury, Eomes, Sox17, FoxA2, relative to high oxygen cultures. The effects extended to the subsequent formation of more mature mesodermal lineages. We observed significant upregulation of cardiomyocyte marker Nkx2.5, and critically a decrease in the number of contaminant pluripotent cells after 12 days using a directed cardiomyocyte protocol. However, the impact of low oxygen preconditioning was to prime human cells for ectodermal lineage commitment during subsequent EB differentiation, with significant upregulation of Nestin and β3-tubulin. Our research demonstrates the importance of oxygen tension control during cell maintenance on the subsequent differentiation of both mouse and human PSCs, and highlights the differential effects.
Collapse
Affiliation(s)
- Kate Fynes
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London , London, United Kingdom
| | | | | | | | | | | |
Collapse
|
28
|
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: 2.6] [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
|
29
|
Wang S, Zhou Y, Andreyev O, Hoyt RF, Singh A, Hunt T, Horvath KA. Overexpression of FABP3 inhibits human bone marrow derived mesenchymal stem cell proliferation but enhances their survival in hypoxia. Exp Cell Res 2014; 323:56-65. [PMID: 24583397 DOI: 10.1016/j.yexcr.2014.02.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/25/2014] [Accepted: 02/16/2014] [Indexed: 11/29/2022]
Abstract
Studying the proliferative ability of human bone marrow derived mesenchymal stem cells in hypoxic conditions can help us achieve the effective regeneration of ischemic injured myocardium. Cardiac-type fatty acid binding protein (FABP3) is a specific biomarker of muscle and heart tissue injury. This protein is purported to be involved in early myocardial development, adult myocardial tissue repair and responsible for the modulation of cell growth and proliferation. We have investigated the role of FABP3 in human bone marrow derived mesenchymal stem cells under ischemic conditions. MSCs from 12 donors were cultured either in standard normoxic or modified hypoxic conditions, and the differential expression of FABP3 was tested by quantitative (RT)PCR and western blot. We also established stable FABP3 expression in MSCs and searched for variation in cellular proliferation and differentiation bioprocesses affected by hypoxic conditions. We identified: (1) the FABP3 differential expression pattern in the MSCs under hypoxic conditions; (2) over-expression of FABP3 inhibited the growth and proliferation of the MSCs; however, improved their survival in low oxygen environments; (3) the cell growth factors and positive cell cycle regulation genes, such as PCNA, APC, CCNB1, CCNB2 and CDC6 were all down-regulated; while the key negative cell cycle regulation genes TP53, BRCA1, CASP3 and CDKN1A were significantly up-regulated in the cells with FABP3 overexpression. Our data suggested that FABP3 was up-regulated under hypoxia; also negatively regulated the cell metabolic process and the mitotic cell cycle. Overexpression of FABP3 inhibited cell growth and proliferation via negative regulation of the cell cycle and down-regulation of cell growth factors, but enhances cell survival in hypoxic or ischemic conditions.
Collapse
Affiliation(s)
- Suna Wang
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yifu Zhou
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oleg Andreyev
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert F Hoyt
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Avneesh Singh
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy Hunt
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keith A Horvath
- Cellular Biology Section, Cardiothoracic Surgery Research Program, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
30
|
Age-Dependent Association between Protein Expression of the Embryonic Stem Cell Marker Cripto-1 and Survival of Glioblastoma Patients. Transl Oncol 2013; 6:732-41. [PMID: 24466376 DOI: 10.1593/tlo.13427] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 08/13/2013] [Accepted: 08/18/2013] [Indexed: 12/26/2022] Open
Abstract
Exploring the re-emergence of embryonic signaling pathways may reveal important information for cancer biology. Nodal is a transforming growth factor-β (TGF-β)-related morphogen that plays a critical role during embryonic development. Nodal signaling is regulated by the Cripto-1 co-receptor and another TGF-β member, Lefty. Although these molecules are poorly detected in differentiated tissues, they have been found in different human cancers. Poor prognosis of glioblastomas justifies the search for novel signaling pathways that can be exploited as potential therapeutic targets. Because our intracranial glioblastoma rat xenograft model has revealed importance of gene ontology categories related to development and differentiation, we hypothesized that increased activity of Nodal signaling could be found in glioblastomas. We examined the gene expressions of Nodal, Cripto-1, and Lefty in microarrays of invasive and angiogenic xenograft samples developed from four patients with glioblastoma. Protein expression was evaluated by immunohistochemistry in 199 primary glioblastomas, and expression levels were analyzed for detection of correlations with available clinical information. Gene expression of Nodal, Lefty, and Cripto-1 was detected in the glioblastoma xenografts. Most patient samples showed significant levels of Cripto-1 detected by immunohistochemistry, whereas only weak to moderate levels were detected for Nodal and Lefty. Most importantly, the higher Cripto-1 scores were associated with shorter survival in a subset of younger patients. These findings suggest for the first time that Cripto-1, an important molecule in developmental biology, may represent a novel prognostic marker and therapeutic target in categories of younger patients with glioblastoma.
Collapse
|
31
|
Wielscher M, Liou W, Pulverer W, Singer CF, Rappaport-Fuerhauser C, Kandioler D, Egger G, Weinhäusel A. Cytosine 5-Hydroxymethylation of the LZTS1 Gene Is Reduced in Breast Cancer. Transl Oncol 2013; 6:715-21. [PMID: 24466374 PMCID: PMC3890706 DOI: 10.1593/tlo.13523] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/06/2013] [Accepted: 09/09/2013] [Indexed: 02/07/2023] Open
Abstract
Change of DNA cytosine methylation (5mC) is an early event in the development of cancer, and the recent discovery of a 5-hydroxymethylated form (5hmC) of cytosine suggests a regulatory epigenetic role that might be different from 5-methylcytosine. Here, we aimed at elucidating the role of 5hmC in breast cancer. To interrogate the 5hmC levels of the leucine zipper, putative tumor suppressor 1 (LZTS1) gene in detail, we analyzed 75 primary breast cancer tissue samples from initial diagnosis and 12 normal breast tissue samples derived from healthy persons. Samples were subjected to 5hmC glucosyltransferase treatment followed by restriction digestion and segment-specific amplification of 11 polymerase chain reaction products. Nine of the 11 5'LZTS1 fragments showed significantly lower (fold change of 1.61-6.01, P < .05) 5hmC content in primary breast cancer tissue compared to normal breast tissue samples. No significant differences were observed for 5mC DNA methylation. Furthermore, both LZTS1 and TET1 mRNA expressions were significantly reduced in tumor samples (n = 75, P < .001, Student's t test), which correlated significantly with 5hmC levels in samples. 5hmC levels in breast cancer tissues were associated with unfavorable histopathologic parameters such as lymph node involvement (P < .05, Student's t test). A decrease of 5hmC levels of LZTS1, a classic tumor suppressor gene known to influence metastasis in breast cancer progression, is correlated to down-regulation of LZTS1 mRNA expression in breast cancer and might epigenetically enhance carcinogenesis. The study provides support for the novel hypothesis that suggests a strong influence of 5hmC on mRNA expression. Finally, one may also consider 5hmC as a new biomarker.
Collapse
Affiliation(s)
- Matthias Wielscher
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Willy Liou
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Walter Pulverer
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| | - Christian F Singer
- Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | | | - Gerda Egger
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Andreas Weinhäusel
- Molecular Diagnostics Unit, Health and Environment Department, Austrian Institute of Technology, Vienna, Austria
| |
Collapse
|
32
|
Effect of oxygen on cardiac differentiation in mouse iPS cells: role of hypoxia inducible factor-1 and Wnt/beta-catenin signaling. PLoS One 2013; 8:e80280. [PMID: 24265804 PMCID: PMC3827186 DOI: 10.1371/journal.pone.0080280] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 10/11/2013] [Indexed: 11/19/2022] Open
Abstract
Background Disturbances in oxygen levels have been found to impair cardiac organogenesis. It is known that stem cells and differentiating cells may respond variably to hypoxic conditions, whereby hypoxia may enhance stem cell pluripotency, while differentiation of multiple cell types can be restricted or enhanced under hypoxia. Here we examined whether HIF-1alpha modulated Wnt signaling affected differentiation of iPS cells into beating cardiomyocytes. Objective We investigated whether transient and sustained hypoxia affects differentiation of cardiomyocytes derived from murine induced pluripotent stem (iPS) cells, assessed the involvement of HIF-1alpha (hypoxia-inducible factor-1alpha) and the canonical Wnt pathway in this process. Methods Embryoid bodies (EBs) derived from iPS cells were differentiated into cardiomyocytes and were exposed either to 24 h normoxia or transient hypoxia followed by a further 13 days of normoxic culture. Results At 14 days of differentiation, 59±2% of normoxic EBs were beating, whilst transient hypoxia abolished beating at 14 days and EBs appeared immature. Hypoxia induced a significant increase in Brachyury and islet-1 mRNA expression, together with reduced troponin C expression. Collectively, these data suggest that transient and sustained hypoxia inhibits maturation of differentiating cardiomyocytes. Compared to normoxia, hypoxia increased HIF-1alpha, Wnt target and ligand genes in EBs, as well as accumulation of HIF-1alpha and beta-catenin in nuclear protein extracts, suggesting involvement of the Wnt/beta-catenin pathway. Conclusion Hypoxia impairs cardiomyocyte differentiation and activates Wnt signaling in undifferentiated iPS cells. Taken together the study suggests that oxygenation levels play a critical role in cardiomyocyte differentiation and suggest that hypoxia may play a role in early cardiogenesis.
Collapse
|
33
|
Bianco C, Castro NP, Baraty C, Rollman K, Held N, Rangel MC, Karasawa H, Gonzales M, Strizzi L, Salomon DS. Regulation of human Cripto-1 expression by nuclear receptors and DNA promoter methylation in human embryonal and breast cancer cells. J Cell Physiol 2013; 228:1174-88. [PMID: 23129342 PMCID: PMC3573215 DOI: 10.1002/jcp.24271] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 10/18/2012] [Indexed: 11/07/2022]
Abstract
Human Cripto-1 (CR-1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR-1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)-1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR-1 gene expression in NTERA-2 human embryonal carcinoma cells and in breast cancer cells. CR-1 expression in NTERA-2 cells was positively regulated by LRH-1 through direct binding to a DR0 element within the CR-1 promoter, while GCNF strongly suppressed CR-1 expression in these cells. In addition, the CR-1 promoter was unmethylated in NTERA-2 cells, while T47D, ZR75-1, and MCF7 breast cancer cells showed high levels of CR-1 promoter methylation and low CR-1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR-1 promoter and reactivated CR-1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR-1 was highly expressed in the majority of human breast tumors, suggesting that CR-1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR-1 gene expression and its critical role in the pathogenesis of human cancer.
Collapse
MESH Headings
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Binding Sites
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Embryonal/genetics
- Carcinoma, Embryonal/metabolism
- Carcinoma, Embryonal/pathology
- Cell Movement
- DNA Methylation/drug effects
- DNA Modification Methylases/antagonists & inhibitors
- DNA Modification Methylases/metabolism
- Decitabine
- Dose-Response Relationship, Drug
- Embryonal Carcinoma Stem Cells/metabolism
- Embryonal Carcinoma Stem Cells/pathology
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Histone Deacetylase Inhibitors/pharmacology
- Humans
- Hydroxamic Acids/pharmacology
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Luciferases/biosynthesis
- Luciferases/genetics
- MCF-7 Cells
- Neoplasm Invasiveness
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Nuclear Receptor Subfamily 6, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism
- Promoter Regions, Genetic
- RNA Interference
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Time Factors
- Tissue Array Analysis
- Transcription, Genetic
- Transfection
- Tretinoin/pharmacology
- Valproic Acid/pharmacology
Collapse
Affiliation(s)
- Caterina Bianco
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nadia P. Castro
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Christina Baraty
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Kelly Rollman
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Natalie Held
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Maria Cristina Rangel
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Hideaki Karasawa
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Monica Gonzales
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Luigi Strizzi
- Children’s Memorial Research Center, Robert H. Lurie Comprehensive Cancer Center Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David S. Salomon
- Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
34
|
Singh RP, Franke K, Wielockx B. Hypoxia-mediated regulation of stem cell fate. High Alt Med Biol 2013; 13:162-8. [PMID: 22994515 DOI: 10.1089/ham.2012.1043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hypoxia-mediated regulation of stem cell fate, or reduced oxygen availability, is a prominent feature during mammalian development and under physiological and pathological conditions in adults. Oxygen-sensing is therefore indispensable as it enables the cells to adapt instantaneously to an inappropriate pO(2). This machinery relies primarily on hypoxia inducible factor (HIF). Moreover, a growing body of evidence proposes that different types of stem cells exist in a very hypoxic microenvironment, which may be beneficial for the maintenance of these cells and ensures continuous replenishment of dead or damaged cells in virtually all tissues of the body. Recent reports have shown that HIF is a critical player in these responses. However, a better understanding of the different HIF-related mechanisms is of utmost importance for the improvement of therapeutic strategies for tissue regeneration as well as hematological malignancies.
Collapse
Affiliation(s)
- Rashim Pal Singh
- Emmy Noether Research Group, Institute of Pathology, University of Technology Dresden, Germany
| | | | | |
Collapse
|
35
|
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: 2.1] [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
|
36
|
Bae D, Mondragon-Teran P, Hernandez D, Ruban L, Mason C, Bhattacharya SS, Veraitch FS. Hypoxia Enhances the Generation of Retinal Progenitor Cells from Human Induced Pluripotent and Embryonic Stem Cells. Stem Cells Dev 2012; 21:1344-55. [DOI: 10.1089/scd.2011.0225] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Daekyeong Bae
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Paul Mondragon-Teran
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Diana Hernandez
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Ludmila Ruban
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Chris Mason
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Shomi S. Bhattacharya
- CABIMER, Isla de la Cartuja, Sevilla, Spain
- UCL-Institute of Ophthalmology, London, United Kingdom
| | - Farlan S. Veraitch
- Department of Biochemical Engineering, The Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
37
|
Rangel MC, Karasawa H, Castro NP, Nagaoka T, Salomon DS, Bianco C. Role of Cripto-1 during epithelial-to-mesenchymal transition in development and cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2188-200. [PMID: 22542493 DOI: 10.1016/j.ajpath.2012.02.031] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 02/13/2012] [Accepted: 02/21/2012] [Indexed: 02/08/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is a critical multistep process that converts epithelial cells to more motile and invasive mesenchymal cells, contributing to body patterning and morphogenesis during embryonic development. In addition, both epithelial plasticity and increased motility and invasiveness are essential for the branching morphogenesis that occurs during development of the mammary gland and during tumor formation, allowing cancer cells to escape from the primary tumor. Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic (EGF/CFC) gene family, together with the transforming growth factor (TGF)-β family ligand Nodal, regulates both cell movement and EMT during embryonic development. During postnatal development, Cripto-1 regulates the branching morphogenesis of the mouse mammary gland and enhances both the invasive and migratory properties of mammary epithelial cells in vitro. Furthermore, transgenic mouse models have shown that Cripto-1 promotes the formation of mammary tumors that display properties of EMT, including the down-regulation of the cell surface adherens junctional protein E-cadherin and the up-regulation of mesenchymal markers, such as vimentin, N-cadherin, and Snail. Interestingly, Cripto-1 is enriched in a subpopulation of embryonal, melanoma, prostate, and pancreatic cancer cells that possess stem-like characteristics. Therefore, Cripto-1 may play a role during developmental EMT, and it may also be involved in the reprogramming of differentiated tumor cells into cancer stem cells through the induction of an EMT program.
Collapse
Affiliation(s)
- Maria C Rangel
- Tumor Growth Factor Section, Laboratory of Cancer Prevention, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
38
|
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: 11.0] [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
|
39
|
Lee KE, Simon MC. From stem cells to cancer stem cells: HIF takes the stage. Curr Opin Cell Biol 2012; 24:232-5. [PMID: 22296771 DOI: 10.1016/j.ceb.2012.01.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 01/06/2012] [Accepted: 01/11/2012] [Indexed: 12/26/2022]
Abstract
Hypoxia, a condition of insufficient oxygen availability, occurs during normal development as well as tumorigenesis. Cellular responses to hypoxia are primarily mediated by hypoxia-inducible factors (HIFs). Recent studies have revealed that dormant hematopoietic stem cells (HSCs) reside within hypoxic regions of the bone marrow and that HIF is a critical player in HSC homeostasis. The functional significance of HIF in maintaining stemness also applies to cancer stem cells in hematological malignancies. These findings indicate that better understanding of the mechanisms underlying HIF functions in stem cells should permit the development of new therapies for tissue regeneration and cancer.
Collapse
Affiliation(s)
- Kyoung Eun Lee
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
| | | |
Collapse
|
40
|
Mondragon-Teran P, Baboo JZ, Mason C, Lye GJ, Veraitch FS. The full spectrum of physiological oxygen tensions and step-changes in oxygen tension affects the neural differentiation of mouse embryonic stem cells. Biotechnol Prog 2011; 27:1700-8. [PMID: 21901864 DOI: 10.1002/btpr.675] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 06/15/2011] [Indexed: 11/06/2022]
Abstract
The beneficial impact of lowering oxygen tension to physiological levels has been demonstrated in a number of stem cell differentiation protocols. The majority of these studies compare normal laboratory oxygen tension with one physiological condition (typically 2-5% O(2) ). In this article, we investigated whether the full spectrum of physiological oxygen tensions (0-20% O(2) ) and step-changes in oxygen tension could enhance the production of neural populations from of embryonic stem cells (ESCs). We used a model system for the conversion of mouse ESCs into cells expressing one neuroectoderm stem cell marker (nestin) and two neural markers (βIII tubulin and microtubule-associated protein (MAP2)). 4-10% O(2) was associated with large increases in the total production of viable cells and the highest number of cells expressing Nestin, βIII tubulin, and MAP2. However, 4-10% O(2) also caused a reduction in the percentage of cells expressing all three markers. Step changes in oxygen tension at the mid-point of the differentiation process affected the total production of viable cells and the percentage of cells expressing all three markers. We found that the initial oxygen tension and the magnitude of the step-change were critical variables. A step increase from 0 to 2% O(2) mid-way through the protocol resulted in the highest percentage of cells expressing βIII tubulin (86.5%). In conclusion, we have demonstrated that the full spectrum of physiological oxygen tensions and step changes in oxygen tension represent a powerful tool for the optimisation of neural differentiation processes.
Collapse
Affiliation(s)
- Paul Mondragon-Teran
- The Advanced Centre for Biochemical Engineering, Dept. of Biochemical Engineering, University College London, Torrington Place, London, UK
| | | | | | | | | |
Collapse
|
41
|
Sapra P, Hooper AT, O'Donnell CJ, Gerber HP. Investigational antibody drug conjugates for solid tumors. Expert Opin Investig Drugs 2011; 20:1131-49. [DOI: 10.1517/13543784.2011.582866] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
42
|
Sarig U, Machluf M. Engineering cell platforms for myocardial regeneration. Expert Opin Biol Ther 2011; 11:1055-77. [DOI: 10.1517/14712598.2011.578574] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
43
|
Lim HJ, Han J, Woo DH, Kim SE, Kim SK, Kang HG, Kim JH. Biochemical and morphological effects of hypoxic environment on human embryonic stem cells in long-term culture and differentiating embryoid bodies. Mol Cells 2011; 31:123-32. [PMID: 21347709 PMCID: PMC3932683 DOI: 10.1007/s10059-011-0016-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 11/06/2010] [Accepted: 11/17/2010] [Indexed: 12/17/2022] Open
Abstract
The mammalian reproductive tract is known to contain 1.5-5.3% oxygen (O(2)), but human embryonic stem cells (hESCs) derived from preimplantation embryos are typically cultured under 21% O(2) tension. The aim of this study was to investigate the effects of O(2) tension on the long-term culture of hESCs and on cell-fate determination during early differentiation. hESCs and embryoid bodies (EBs) were grown under different O(2) tensions (3, 12, and 21% O(2)). The expression of markers associated with pluripotency, embryonic germ layers, and hypoxia was analyzed using RTPCR, immunostaining, and Western blotting. Proliferation, apoptosis, and chromosomal aberrations were examined using BrdU incorporation, caspase-3 immunostaining, and karyotype analysis, respectively. Structural and morphological changes of EBs under different O(2) tensions were comparatively examined using azan- and hematoxylineosin staining, and scanning and transmission electron microscopy. Mild hypoxia (12% O(2)) increased the number of cells expressing Oct4/Nanog and reduced BrdU incorporation and aneuploidy. The percentage of cells positive for active caspase-3, which was high during normoxia (21% O(2)), gradually decreased when hESCs were continuously cultured under mild hypoxia. EBs subjected to hypoxia (3% O(2)) exhibited well-differentiated microvilli on their surface, secreted high levels of collagen, and showed enhanced differentiation into primitive endoderm. These changes were associated with increased expression of Foxa2, Sox17, AFP, and GATA4 on the EB periphery. Our data suggest that mild hypoxia facilitates the slow mitotic division of hESCs in long-term culture and reduces the frequency of chromosomal abnormalities and apoptosis. In addition, hypoxia promotes the differentiation of EBs into extraembryonic endoderm.
Collapse
Affiliation(s)
- Hee-Joung Lim
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
- Biomedical Laboratory, Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Sungnam 461-713, Korea
| | - Jiyou Han
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| | - Dong-Hun Woo
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| | - Sung-Eun Kim
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| | - Suel-Kee Kim
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| | - Hee-Gyoo Kang
- Biomedical Laboratory, Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Sungnam 461-713, Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cell Biology, Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Korea
| |
Collapse
|
44
|
Abstract
IMPORTANCE OF THE FIELD Emerging evidence has clearly implicated an inappropriate activation of embryonic regulatory genes during cell transformation in adult tissues. An example of such a case is the embryonic gene Cripto-1. Cripto-1 is critical for embryonic development and is considered a marker of undifferentiated embryonic stem cells. Critpo-1 is expressed at low levels in adult tissues, but is re-expressed at a high frequency in a number of different types of human carcinomas, therefore, representing an attractive therapeutic target in cancer. AREA COVERED IN THIS REVIEW This review surveys different approaches that have been used to target Cripto-1 in cancer as reflected by the relevant patent literature as well as peer-reviewed publications. Potential involvement and targeting of Cripto-1 in neurodegenerative and degenerative muscle diseases are also discussed. WHAT THE READER WILL GAIN The reader will gain an overview of different mAbs, vaccines or oligonucleotides antisense targeting Cripto-1. A humanized anti-Cripto-1 antibody is currently being tested in a Phase I clinical trial in cancer patients. TAKE HOME MESSAGE Targeting Cripto-1 in human tumors has the potential to eliminate not only differentiated cancer cells but also destroy an undifferentiated subpopulation of cancer cells with stem-like characteristics that support tumor initiation and self-renewal.
Collapse
Affiliation(s)
- Caterina Bianco
- National Cancer Institute, National Institutes of Health, Mammary Biology & Tumorigenesis Laboratory, Bethesda, MD 20892, USA.
| | | |
Collapse
|
45
|
Bianco C, Rangel MC, Castro NP, Nagaoka T, Rollman K, Gonzales M, Salomon DS. Role of Cripto-1 in stem cell maintenance and malignant progression. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:532-40. [PMID: 20616345 DOI: 10.2353/ajpath.2010.100102] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cripto-1 is critical for early embryonic development and, together with its ligand Nodal, has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Like other embryonic genes, Cripto-1 performs important roles in the formation and progression of several types of human tumors, stimulating cell proliferation, migration, epithelial to mesenchymal transition, and tumor angiogenesis. Several studies have demonstrated that cell fate regulation during embryonic development and cell transformation during oncogenesis share common signaling pathways, suggesting that uncontrolled activation of embryonic signaling pathways might drive cell transformation and tumor progression in adult tissues. Here we review our current understanding of how Cripto-1 controls stem cell biology and how it integrates with other major embryonic signaling pathways. Because many cancers are thought to derive from a subpopulation of cancer stem-like cells, which may re-express embryonic genes, Cripto-1 signaling may drive tumor growth through the generation or expansion of tumor initiating cells bearing stem-like characteristics. Therefore, the Cripto-1/Nodal signaling may represent an attractive target for treatment in cancer, leading to the elimination of undifferentiated stem-like tumor initiating cells.
Collapse
Affiliation(s)
- Caterina Bianco
- Mammary Biology and Tumorigenesis Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
de Castro NP, Rangel MC, Nagaoka T, Salomon DS, Bianco C. Cripto-1: an embryonic gene that promotes tumorigenesis. Future Oncol 2010; 6:1127-42. [DOI: 10.2217/fon.10.68] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several studies have shown that cell fate regulation during embryonic development and oncogenic transformation share common regulatory mechanisms and signaling pathways. Indeed, an embryonic gene member of the EGF–Cripto-1/FRL1/Cryptic family, Cripto-1, has been implicated in embryogenesis and in carcinogenesis. Cripto-1 together with the TGF-β ligand Nodal is a key regulator of embryonic development and is a marker of undifferentiated human and mouse embryonic stem cells. While Cripto-1 expression is very low in normal adult tissues, Cripto-1 is re-expressed at high levels in several different human tumors, modulating cancer cell proliferation, migration, epithelial-to-mesenchymal transition and stimulating tumor angiogenesis. Therefore, inhibition of Cripto-1 expression using blocking antibodies or antisense expression vectors might be a useful modality not only to target fully differentiated cancer cells but also to target a subpopulation of tumor cells with stem-like characteristics.
Collapse
Affiliation(s)
- Nadia Pereira de Castro
- Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bldg 37 Room 1112, Bethesda, MD 20892, USA
| | - Maria Cristina Rangel
- Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bldg 37 Room 1112, Bethesda, MD 20892, USA
| | - Tadahiro Nagaoka
- Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bldg 37 Room 1112, Bethesda, MD 20892, USA
| | - David S Salomon
- Mammary Biology & Tumorigenesis Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Bldg 37 Room 1112, Bethesda, MD 20892, USA
| | | |
Collapse
|