1
|
Xiao J, Ang JW, Zhong X, Wong DCP, T T, Yow I, Lee CJM, Foo RSY, Kanchanawong P, Low BC. Coordination of Focal Adhesion Nanoarchitecture and Dynamics in Mechanosensing for Cardiomyoblast Differentiation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4463-4479. [PMID: 39778877 PMCID: PMC11758775 DOI: 10.1021/acsami.4c15459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Focal adhesions (FAs) are force-bearing multiprotein complexes, whose nanoscale organization and signaling are essential for cell growth and differentiation. However, the specific organization of FA components to exert spatiotemporal activation of FA proteins for force sensing and transduction remains unclear. In this study, we unveil the intricacies of FA protein nanoarchitecture and that its dynamics are coordinated by a molecular scaffold protein, BNIP-2, to initiate downstream signal transduction for cardiomyoblast differentiation. Within the FAs, BNIP-2 regulates the nano-organization of focal adhesion kinase (FAK), and the dynamics of FAK, paxillin, and vinculin. Depletion of BNIP-2 resulted in altered focal adhesion numbers and sizes per cell, reduced traction force, and decreased FA sensitivity for mechanosensing. At the molecular level, the loss of BNIP-2 disrupted the FAK-paxillin signaling axis, where FAK inhibition reproduces the effects of BNIP-2 loss by impairing the phosphorylation of both FAK and paxillin. Mechanistically, BNIP-2 preferentially binds to constitutively active FAK and acts as a molecular scaffold to mediate interactions between FAK and paxillin and between paxillin and vinculin. We have validated BNIP-2's role in the FAK-paxillin signaling axis in human embryonic stem cells (hESC). Furthermore, we showed that depletion of BNIP-2 resulted in changes in signature gene targets at the cardiac progenitor stage of differentiation. In summary, we showed that the intricate interplay of FA nanoarchitecture and dynamics, governed by BNIP-2, is crucial for force transduction and biochemical signaling in driving cardiomyoblast differentiation.
Collapse
Affiliation(s)
- Jingwei Xiao
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Jing Wen Ang
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Xueying Zhong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Darren Chen Pei Wong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biological Sciences, National University
of Singapore, Singapore 117558, Singapore
| | - Thivakar T
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Ivan Yow
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
| | - Chang Jie Mick Lee
- Institute
of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Cardiovascular
Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, Singapore 117599, Singapore
| | - Roger S-Y Foo
- Institute
of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
- Department
of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Cardiovascular
Metabolic Disease Translational Research Programme, National University Health System, Centre for Translational Medicine, Singapore 117599, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biomedical Engineering, National University
of Singapore, Singapore 117583, Singapore
| | - Boon Chuan Low
- Mechanobiology
Institute Singapore, National University
of Singapore, Singapore 117411, Singapore
- Department
of Biological Sciences, National University
of Singapore, Singapore 117558, Singapore
- NUS
College, National University of Singapore, Singapore 138593, Singapore
| |
Collapse
|
2
|
Casarella S, Ferla F, Di Francesco D, Canciani E, Rizzi M, Boccafoschi F. Focal Adhesion's Role in Cardiomyocytes Function: From Cardiomyogenesis to Mechanotransduction. Cells 2024; 13:664. [PMID: 38667279 PMCID: PMC11049660 DOI: 10.3390/cells13080664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Mechanotransduction refers to the ability of cells to sense mechanical stimuli and convert them into biochemical signals. In this context, the key players are focal adhesions (FAs): multiprotein complexes that link intracellular actin bundles and the extracellular matrix (ECM). FAs are involved in cellular adhesion, growth, differentiation, gene expression, migration, communication, force transmission, and contractility. Focal adhesion signaling molecules, including Focal Adhesion Kinase (FAK), integrins, vinculin, and paxillin, also play pivotal roles in cardiomyogenesis, impacting cell proliferation and heart tube looping. In fact, cardiomyocytes sense ECM stiffness through integrins, modulating signaling pathways like PI3K/AKT and Wnt/β-catenin. Moreover, FAK/Src complex activation mediates cardiac hypertrophic growth and survival signaling in response to mechanical loads. This review provides an overview of the molecular and mechanical mechanisms underlying the crosstalk between FAs and cardiac differentiation, as well as the role of FA-mediated mechanotransduction in guiding cardiac muscle responses to mechanical stimuli.
Collapse
Affiliation(s)
- Simona Casarella
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Federica Ferla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Dalila Di Francesco
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Elena Canciani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Manuela Rizzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| |
Collapse
|
3
|
Xu F, Zheng Z, Yao M, Zhu F, Shen T, Li J, Zhu C, Yang T, Shao M, Wan Z, Fang C. A regulatory mechanism of a stepwise osteogenesis-mimicking decellularized extracellular matrix on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. J Mater Chem B 2022; 10:6171-6180. [PMID: 35766339 DOI: 10.1039/d2tb00721e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A cell-derived decellularized extracellular matrix (dECM) plays a vital role in controlling cell functions because of its similarity to the in vivo microenvironment. In the process of stem cell differentiation, the composition of the dECM is not constant but is dynamically remolded. However, there is little information regarding the dynamic regulation by the dECM of the osteogenic differentiation of stem cells. Herein, four types of stepwise dECMs (0, 7, 14, and 21 d-ECM) were prepared from bone marrow-derived mesenchymal stem cells (BMSCs) undergoing osteogenic differentiation for 0, 7, 14, and 21 days after decellularization. In vitro experiments were designed to study the regulation of BMSC osteogenesis by dECMs. The results showed that all the dECMs could support the activity and proliferation of BMSCs but had different effects on their osteogenic differentiation. The 14d-ECM promoted the osteogenesis of BMSCs significantly compared with the other dECMs. Proteomic analysis demonstrated that the composition of dECMs changed over time. The 14d ECM had higher amounts of collagen type IV alpha 2 chain (COL4A2) than the other dECMs. Furthermore, COL4A2 was obviously enriched in the activated focal adhesion kinase (FAK)/phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways. Thus, the 14d-ECM could promote the osteogenic differentiation of BMSCs, which might be related to the high content of COL4A2 in the 14d-ECM by activating the FAK/PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Fei Xu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Ziran Zheng
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Mianfeng Yao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Feiya Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Ting Shen
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Jiang Li
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Chao Zhu
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| | - Tianru Yang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Mengying Shao
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.
| | - Zicheng Wan
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Changyun Fang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China. .,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Li K, Wang X, Fan C, Wu C, Li S, Liu H. Tanshinone IIA promotes cardiac differentiation and improves cell motility by modulating the Wnt/β‑catenin signaling pathway. Mol Med Rep 2020; 22:1839-1846. [PMID: 32582982 PMCID: PMC7411398 DOI: 10.3892/mmr.2020.11272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Although the cardiovascular pharmacological actions of Tanshinone IIA (TanIIA) have been extensively studied, research on its roles in cardiac regeneration is still insufficient. The present study employed the cardiac myoblast cell line H9c2 to evaluate the possible roles of TanIIA in cardiac regeneration. It was found that certain concentration of TanIIA inhibited cell proliferation by suppressing the expression of proteins related to the cell cycle [cyclin dependent kinase (CDK)4, CDK6 and cyclin D1] and proliferation [c-Myc, octamer-binding transcription factor 4 (Oct4) and proliferating cell nuclear antigen (PCNA)] without inducing apoptosis. In this process, the expression of cardiac troponin in the treated cells was significantly increased and the migration of the treated cells toward the wound area was significantly enhanced. Meanwhile, TanIIA inhibited the canonical signaling pathway through increasing the expression of glycogen synthase kinase 3β (GSK-3β) and adenomatous polyposis coli (APC) and increased the expression of Wnt11 and Wnt5a in the noncanonical Wnt signaling pathway. Following β-catenin agonist WAY-262611 intervention, the effect of TanIIA on the promotion of cardiac differentiation and improved cell migration was significantly reduced. In conclusion, it was hypothesized that TanIIA could promote cardiac differentiation and improve cell motility by modulating the Wnt/β-catenin signaling pathway. These results suggest that TanIIA may play beneficial roles in myocardial regeneration following stem cell transplantation.
Collapse
Affiliation(s)
- Kun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xiuyan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Chenxing Fan
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Chunxia Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Shizheng Li
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Hua Liu
- Institute of Eyes, Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
5
|
Tsui JH, Ostrovsky-Snider NA, Yama DMP, Donohue JD, Choi JS, Chavanachat R, Larson JD, Murphy AR, Kim DH. Conductive Silk-Polypyrrole Composite Scaffolds with Bioinspired Nanotopographic Cues for Cardiac Tissue Engineering. J Mater Chem B 2018; 6:7185-7196. [PMID: 31448124 PMCID: PMC6708520 DOI: 10.1039/c8tb01116h] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We report on the development of bioinspired cardiac scaffolds made from electroconductive acid-modified silk fibroin-poly(pyrrole) (AMSF+PPy) substrates patterned with nanoscale ridges and grooves reminiscent of native myocardial extracellular matrix (ECM) topography to enhance the structural and functional properties of cultured human pluripotent stem cells (hPSC)-derived cardiomyocytes. Nanopattern fidelity was maintained throughout the fabrication and functionalization processes, and no loss in conductive behavior occurred due to the presence of the nanotopographical features. AMSF+PPy substrates were biocompatible and stable, maintaining high cell viability over a 21-day culture period while displaying no signs of PPy delamination. The presence of anisotropic topographical cues led to increased cellular organization and sarcomere development, and electroconductive cues promoted a significant improvement in the expression and polarization of connexin 43 (Cx43), a critical regulator of cell-cell electrical coupling. The combination of biomimetic topography and electroconductivity also increased the expression of genes that encode key proteins involved in regulating the contractile and electrophysiological function of mature human cardiac tissue.
Collapse
Affiliation(s)
- Jonathan H. Tsui
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - David M. P. Yama
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jordan D. Donohue
- Department of Chemistry, Western Washington University, Bellingham, WA, USA
| | - Jong Seob Choi
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - Jesse D. Larson
- Department of Chemistry, Western Washington University, Bellingham, WA, USA
| | - Amanda R. Murphy
- Department of Chemistry, Western Washington University, Bellingham, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Li T, He Z, Zhang X, Tian M, Jiang K, Cheng G, Wang Y. The status of MAPK cascades contributes to the induction and activation of Gata4 and Nkx2.5 during the stepwise process of cardiac differentiation. Cell Signal 2018; 54:17-26. [PMID: 30471465 DOI: 10.1016/j.cellsig.2018.11.019] [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: 10/11/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 12/28/2022]
Abstract
Cardiac differentiation in vitro is a complex, stepwise process that is rigidly governed by a subset of transcription factors and signaling cascades. In this study, we investigated the cooperation of cardiac-specific transcription factors Gata4 and Nkx2.5, as well as mitogen-activated protein kinase (MAPK) cascades. P19 embryonic carcinoma cells were induced into spontaneously beating cardiomyocytes utilizing a two-step protocol that comprised an early stage and a late stage of differentiation. During early-stage differentiation in suspension culture, P19 cells aggregated to form embryoid bodies (EBs), and the Gata4 and Nkx2.5 genes were induced. However, Gata4 expressed at the early stage of differentiation was incapable of activating downstream gene expression, as it was localized in the cytoplasm and prone to degradation. After EBs were plated for late-stage differentiation in adherent culture, the MAPK cascades were highly activated and contributed to the activation of Gata4 and Nkx2.5. Specifically, we revealed that p38 signaling participated in regulating the localization and stabilization of Gata4 and Nkx2.5. Additionally, the JNK cascade regulated late-stage cardiac differentiation; JNK kinase reduced Gata4 stabilization and conversely alleviated Nkx2.5 degradation by direct interaction and phosphorylation of Nkx2.5. Finally, we found that the C-terminal domain of Nkx2.5 was required for its stabilization under conditions of oxidative stress and JNK activation. Overall, our results indicated that the induction and activation of Gata4 and Nkx2.5 during early- and late-stage cardiac differentiation was closely associated with the function of the MAPK signaling cascades.
Collapse
Affiliation(s)
- Tao Li
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China.
| | - Zezhao He
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Xia Zhang
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Mei Tian
- School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| | - Kesheng Jiang
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Guanchang Cheng
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China
| | - Yunlong Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
7
|
Wang Y, Terrell AM, Riggio BA, Anand D, Lachke SA, Duncan MK. β1-Integrin Deletion From the Lens Activates Cellular Stress Responses Leading to Apoptosis and Fibrosis. Invest Ophthalmol Vis Sci 2017; 58:3896-3922. [PMID: 28763805 PMCID: PMC5539801 DOI: 10.1167/iovs.17-21721] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/30/2017] [Indexed: 12/18/2022] Open
Abstract
Purpose Previous research showed that the absence of β1-integrin from the mouse lens after embryonic day (E) 13.5 (β1MLR10) leads to the perinatal apoptosis of lens epithelial cells (LECs) resulting in severe microphthalmia. This study focuses on elucidating the molecular connections between β1-integrin deletion and this phenotype. Methods RNA sequencing was performed to identify differentially regulated genes (DRGs) in β1MLR10 lenses at E15.5. By using bioinformatics analysis and literature searching, Egr1 (early growth response 1) was selected for further study. The activation status of certain signaling pathways (focal adhesion kinase [FAK]/Erk, TGF-β, and Akt signaling) was studied via Western blot and immunohistochemistry. Mice lacking both β1-integrin and Egr1 genes from the lenses were created (β1MLR10/Egr1-/-) to study their relationship. Results RNA sequencing identified 120 DRGs that include candidates involved in the cellular stress response, fibrosis, and/or apoptosis. Egr1 was investigated in detail, as it mediates cellular stress responses in various cell types, and is recognized as an upstream regulator of numerous other β1MLR10 lens DRGs. In β1MLR10 mice, Egr1 levels are elevated shortly after β1-integrin loss from the lens. Further, pErk1/2 and pAkt are elevated in β1MLR10 LECs, thus providing the potential signaling mechanism that causes Egr1 upregulation in the mutant. Indeed, deletion of Egr1 from β1MLR10 lenses partially rescues the microphthalmia phenotype. Conclusions β1-integrin regulates the appropriate levels of Erk1/2 and Akt phosphorylation in LECs, whereas its deficiency results in the overexpression of Egr1, culminating in reduced cell survival. These findings provide insight into the molecular mechanism underlying the microphthalmia observed in β1MLR10 mice.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Anne M. Terrell
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Brittany A. Riggio
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Melinda K. Duncan
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
8
|
Abbey D, Seshagiri PB. Ascorbic acid-mediated enhanced cardiomyocyte differentiation of mouse ES-cells involves interplay of DNA methylation and multiple-signals. Differentiation 2017; 96:1-14. [PMID: 28554048 DOI: 10.1016/j.diff.2017.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 02/23/2017] [Accepted: 04/25/2017] [Indexed: 01/10/2023]
Abstract
Embryonic stem cells (ES-cells) provide a good model system to study lineage-specific differentiation. Though, the differentiation of ES-cells to cardiomyocytes is documented, a clear understanding of the molecular mechanism of differentiation and improved functional-differentiation efficiency are yet to be achieved. In this regard, ascorbic acid (Aa) is shown to be one of the effective cardiac inducers in ES-cells. But, its mechanism is poorly understood. We therefore, investigated the mechanism of Aa-mediated cardiomyocyte differentiation of ES-cells. Here, we describe the potential involvement of epigenetic (DNA methylation) as well as integrin- and Erk- signaling systems during cardiomyocyte differentiation. Transgenic GS-2 ES-cells and wild-type D3 ES-cells were differentiated to cardiomyocytes, in the presence or absence of Aa and with or without inhibitors of Erk-, collagen- and integrin- pathways. At specific time points, differentiated states of ES-cells were scored by gene expression analyses and the proportion of functional cTnI+ cardiomyocytes. DNA methylation changes of Isl-1, BMP-2, GATA-4 and α-MHC in cardiogenic cells, following stimulation with Aa, were analyzed by using methylation specific PCR (MSP). We observed that Aa, when applied in initial phase of ES-cell differentiation, consistently enhanced cardiac differentiation (99%) over that observed during spontaneous differentiation (70%). This was associated with enhanced expressions of cardiogenesis-associated genes. A two-fold increase in cTnI+ cells was observed, with appropriate myofibril arrangement. The observed effect of Aa was due to enhanced collagen and integrin signaling, coupled with a high p-ERK1/2 expression, downstream. Besides, the involvement of DNA methylation in regulating the expression of cardiac genes i.e., Isl-1 and α-MHC was also observed. Overall, this study, for the first time, demonstrates that Aa-mediated cardiac enhancement is brought about, mechanistically, through the interplay of epigenetic changes in DNA methylation of cardiac genes (Isl-1 and α-MHC) and integrin signaling system.
Collapse
Affiliation(s)
- Deepti Abbey
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
9
|
Li W, Chen L, Chen Z, Wu L, Feng J, Wang F, Shoff L, Li X, Donly KJ, MacDougall M, Chen S. Dentin sialoprotein facilitates dental mesenchymal cell differentiation and dentin formation. Sci Rep 2017; 7:300. [PMID: 28331230 PMCID: PMC5428264 DOI: 10.1038/s41598-017-00339-w] [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: 09/13/2016] [Accepted: 02/22/2017] [Indexed: 01/09/2023] Open
Abstract
Dentin sialoprotein (DSP) is a dentin extracellular matrix protein. It is involved in dental mesenchymal cell lineages and dentin formation through regulation of its target gene expression. DSP mutations cause dentin genetic diseases. However, mechanisms of DSP in controlling dental mesenchymal cell differentiation are unknown. Using DSP as bait, we screened a protein library from mouse odontoblastic cells and found that DSP is a ligand and binds to cell surface receptor, occludin. Further study identified that the C-terminal DSP domainaa 363–458 interacts with the occludin extracellular loop 2aa 194–241. The C-terminal DSP domain induced phosphorylation of occludin Ser490 and focal adhesion kinase (FAK) Ser722 and Tyr576. Coexpression of DSP, occludin and FAK was detected in dental mesenchymal cells during tooth development. Occludin physically interacts with FAK, and occludin and FAK phosphorylation can be blocked by DSP and occludin antibodies. This DSP domain facilitates dental mesenchymal cell differentiation and mineralization. Furthermore, transplantation and pulp-capping procedures revealed that this DSP domain induces endogenous dental pulp mesenchymal cell proliferation, differentiation and migration, while stimulating blood vessel proliferation. This study elucidates the mechanism of DSP in dental mesenchymal lineages and implies that DSP may serve as a therapeutic agent for dentin-pulp complex regeneration in dental caries.
Collapse
Affiliation(s)
- Wentong Li
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States.,Department of Pathology, Weifang Medical University, Weifang, Shandong Province, 261053, China
| | - Lei Chen
- Department of Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350108, China
| | - Zhuo Chen
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Lian Wu
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Junsheng Feng
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Feng Wang
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Lisa Shoff
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Xin Li
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Kevin J Donly
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States
| | - Mary MacDougall
- Department of Oral/Maxillofacial Surgery, University of Alabama at Birmingham School of Dentistry, Birmingham, Alabama, 35294-0007, United States
| | - Shuo Chen
- Department of Developmental Dentistry, the University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3700, United States.
| |
Collapse
|
10
|
Xu HY, Nie EM, Deng G, Lai LZ, Sun FY, Tian H, Fang FC, Zou YG, Wu BL, Ou-Yang J. Periostin is essential for periodontal ligament remodeling during orthodontic treatment. Mol Med Rep 2017; 15:1800-1806. [DOI: 10.3892/mmr.2017.6200] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/09/2016] [Indexed: 11/06/2022] Open
|
11
|
Li XL, Zeng D, Chen Y, Ding L, Li WJ, Wei T, Ou DB, Yan S, Wang B, Zheng QS. Role of alpha- and beta-adrenergic receptors in cardiomyocyte differentiation from murine-induced pluripotent stem cells. Cell Prolif 2016; 50. [PMID: 27790820 DOI: 10.1111/cpr.12310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/13/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a promising source of cells for regenerative heart disease therapies, but progress towards their use has been limited by their low differentiation efficiency and high cellular heterogeneity. Previous studies have demonstrated expression of adrenergic receptors (ARs) in stem cells after differentiation; however, roles of ARs in fate specification of stem cells, particularly in cardiomyocyte differentiation and development, have not been characterized. MATERIALS AND METHODS Murine-induced pluripotent stem cells (miPSCs) were cultured in hanging drops to form embryoid bodies, cells of which were then differentiated into cardiomyocytes. To determine whether ARs regulated miPSC differentiation into cardiac lineages, effects of the AR agonist, epinephrine (EPI), on miPSC differentiation and underlying signalling mechanisms, were evaluated. RESULTS Treatment with EPI, robustly enhanced miPSC cardiac differentiation, as indicated by increased expression levels of cardiac-specific markers, GATA4, Nkx2.5 and Tnnt2. Although β-AR signalling is the foremost signalling pathway in cardiomyocytes, EPI-enhanced cardiac differentiation depended more on α-AR signalling than β-AR signalling. In addition, selective activation of α1 -AR signalling with specific agonists induced vigorous cardiomyocyte differentiation, whereas selective activation of α2 - or β-AR signalling induced no or less differentiation, respectively. EPI- and α1 -AR-dependent cardiomyocyte differentiation from miPSCs occurred through specific promotion of CPC proliferation via the MEK-ERK1/2 pathway and regulation of miPS cell-cycle progression. CONCLUSIONS These results demonstrate that activation of ARs, particularly of α1 -ARs, promoted miPSC differentiation into cardiac lineages via MEK-ERK1/2 signalling.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Chen
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Department of Emergency, Chinese PLA No.401 Hospital, Qingdao, 266071, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Wen-Ju Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Dong-Bo Ou
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Song Yan
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Bin Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Qiang-Sun Zheng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
12
|
Shekhar A, Lin X, Liu FY, Zhang J, Mo H, Bastarache L, Denny JC, Cox NJ, Delmar M, Roden DM, Fishman GI, Park DS. Transcription factor ETV1 is essential for rapid conduction in the heart. J Clin Invest 2016; 126:4444-4459. [PMID: 27775552 DOI: 10.1172/jci87968] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 09/15/2016] [Indexed: 01/12/2023] Open
Abstract
Rapid impulse propagation in the heart is a defining property of pectinated atrial myocardium (PAM) and the ventricular conduction system (VCS) and is essential for maintaining normal cardiac rhythm and optimal cardiac output. Conduction defects in these tissues produce a disproportionate burden of arrhythmic disease and are major predictors of mortality in heart failure patients. Despite the clinical importance, little is known about the gene regulatory network that dictates the fast conduction phenotype. Here, we have used signal transduction and transcriptional profiling screens to identify a genetic pathway that converges on the NRG1-responsive transcription factor ETV1 as a critical regulator of fast conduction physiology for PAM and VCS cardiomyocytes. Etv1 was highly expressed in murine PAM and VCS cardiomyocytes, where it regulates expression of Nkx2-5, Gja5, and Scn5a, key cardiac genes required for rapid conduction. Mice deficient in Etv1 exhibited marked cardiac conduction defects coupled with developmental abnormalities of the VCS. Loss of Etv1 resulted in a complete disruption of the normal sodium current heterogeneity that exists between atrial, VCS, and ventricular myocytes. Lastly, a phenome-wide association study identified a link between ETV1 and bundle branch block and heart block in humans. Together, these results identify ETV1 as a critical factor in determining fast conduction physiology in the heart.
Collapse
|
13
|
Ishida H, Saba R, Kokkinopoulos I, Hashimoto M, Yamaguchi O, Nowotschin S, Shiraishi M, Ruchaya P, Miller D, Harmer S, Poliandri A, Kogaki S, Sakata Y, Dunkel L, Tinker A, Hadjantonakis AK, Sawa Y, Sasaki H, Ozono K, Suzuki K, Yashiro K. GFRA2 Identifies Cardiac Progenitors and Mediates Cardiomyocyte Differentiation in a RET-Independent Signaling Pathway. Cell Rep 2016; 16:1026-1038. [PMID: 27396331 PMCID: PMC4967477 DOI: 10.1016/j.celrep.2016.06.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/22/2016] [Accepted: 06/10/2016] [Indexed: 12/14/2022] Open
Abstract
A surface marker that distinctly identifies cardiac progenitors (CPs) is essential for the robust isolation of these cells, circumventing the necessity of genetic modification. Here, we demonstrate that a Glycosylphosphatidylinositol-anchor containing neurotrophic factor receptor, Glial cell line-derived neurotrophic factor receptor alpha 2 (Gfra2), specifically marks CPs. GFRA2 expression facilitates the isolation of CPs by fluorescence activated cell sorting from differentiating mouse and human pluripotent stem cells. Gfra2 mutants reveal an important role for GFRA2 in cardiomyocyte differentiation and development both in vitro and in vivo. Mechanistically, the cardiac GFRA2 signaling pathway is distinct from the canonical pathway dependent on the RET tyrosine kinase and its established ligands. Collectively, our findings establish a platform for investigating the biology of CPs as a foundation for future development of CP transplantation for treating heart failure.
Collapse
Affiliation(s)
- Hidekazu Ishida
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Department of Paediatrics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Rie Saba
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ioannis Kokkinopoulos
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Masakazu Hashimoto
- Laboratory for Embryogenesis, Osaka University Graduate School of Frontier Biosciences, Osaka 565-0871, Japan
| | - Osamu Yamaguchi
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Sonja Nowotschin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Manabu Shiraishi
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Prashant Ruchaya
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Centre of Human and Aerospace Physiological Sciences, School of Biomedical Sciences, King's College, London, SE1 1UL, UK
| | - Duncan Miller
- Cardiac Electrophysiology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Stephen Harmer
- Cardiac Electrophysiology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ariel Poliandri
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Shigetoyo Kogaki
- Department of Paediatrics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Leo Dunkel
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Andrew Tinker
- Cardiac Electrophysiology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Osaka University Graduate School of Frontier Biosciences, Osaka 565-0871, Japan
| | - Keiichi Ozono
- Department of Paediatrics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Ken Suzuki
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kenta Yashiro
- Centre for Endocrinology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
14
|
Ou D, Wang Q, Huang Y, Zeng D, Wei T, Ding L, Li X, Zheng Q, Jin Y. Co-culture with neonatal cardiomyocytes enhances the proliferation of iPSC-derived cardiomyocytes via FAK/JNK signaling. BMC DEVELOPMENTAL BIOLOGY 2016; 16:11. [PMID: 27141946 PMCID: PMC4855360 DOI: 10.1186/s12861-016-0112-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/26/2016] [Indexed: 01/26/2023]
Abstract
Background We previously reported that the pluripotent stem cells can differentiate into cardiomyocytes (CMs) by co-culture with neonatal CMs (NCMs) in vitro. However, the involving mechanism is not clear. Methods Mouse induced pluripotent stem cells (iPSCs) were cultured in hanging drops to form embryoid bodies (EBs) and to induce myocardial differentiation. Co-culture of EBs and NCMs was established in a transwell insert system, while EBs grown alone in the wells were used as controls. Results Co-culture with NCMs markedly increased the generation of functional CMs from iPSCs. The focal adhesion kinase (FAK) phosphorylation, and c-Jun N-terminal kinase (JNK) phosphorylation in co-culture were higher than that in EBs grown alone. Treating FAK small interfering RNA (FAK siRNA) or specific inhibitor for JNK (SP600125) to iPSCs significantly reduced the phosphorylation of JNK and the expressions of Mef2c and Bcl-2. The expressions of cTnT and MLC-2V were also decreased. Our results revealed that co-culture with NCMs significantly enhance the differentiation ability of iPSCs by increasing Mef2c and Bcl-2 expressions concomitantly with a marked augment on cell proliferation through JNK signaling pathways. Conclusions These findings indicated that co-culture of EBs with NCMs induces genes expressed in a mature pattern and stimulates the proliferation of iPSC-derived CMs (iPS-CMs) by activating FAK/JNK signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0112-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dongbo Ou
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.,Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qi Wang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Yanjin Huang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Xiaoli Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qiangsun Zheng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China. .,Research and Development Center for Tissue Engineering, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
15
|
Shoni M, Lui KO, Vavvas DG, Muto MG, Berkowitz RS, Vlahos N, Ng SW. Protein kinases and associated pathways in pluripotent state and lineage differentiation. Curr Stem Cell Res Ther 2015; 9:366-87. [PMID: 24998240 DOI: 10.2174/1574888x09666140616130217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/07/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023]
Abstract
Protein kinases (PKs) mediate the reversible conversion of substrate proteins to phosphorylated forms, a key process in controlling intracellular signaling transduction cascades. Pluripotency is, among others, characterized by specifically expressed PKs forming a highly interconnected regulatory network that culminates in a finely-balanced molecular switch. Current high-throughput phosphoproteomic approaches have shed light on the specific regulatory PKs and their function in controlling pluripotent states. Pluripotent cell-derived endothelial and hematopoietic developments represent an example of the importance of pluripotency in cancer therapeutics and organ regeneration. This review attempts to provide the hitherto known kinome profile and the individual characterization of PK-related pathways that regulate pluripotency. Elucidating the underlying intrinsic and extrinsic signals may improve our understanding of the different pluripotent states, the maintenance or induction of pluripotency, and the ability to tailor lineage differentiation, with a particular focus on endothelial cell differentiation for anti-cancer treatment, cell-based tissue engineering, and regenerative medicine strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu-Wing Ng
- 221 Longwood Avenue, BLI- 449A, Boston MA 02115, USA.
| |
Collapse
|
16
|
Nuche-Berenguer B, Moreno P, Jensen RT. Elucidation of the roles of the Src kinases in pancreatic acinar cell signaling. J Cell Biochem 2015; 116:22-36. [PMID: 25079913 PMCID: PMC4229413 DOI: 10.1002/jcb.24895] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/25/2014] [Indexed: 12/13/2022]
Abstract
Recent studies report the Src-family kinases (SFK's) are important in a number of physiological and pathophysiological responses of pancreatic acinar cells (pancreatitis, growth, apoptosis); however, the role of SFKs in various signaling cascades important in mediating these cell functions is either not investigated or unclear. To address this we investigated the action of SFKs in these signaling cascades in rat pancreatic acini by modulating SFK activity using three methods: adenovirus-induced expression of an inactive dominant-negative CSK (Dn-CSK-Advirus) or wild-type CSK (Wt-CSK-Advirus), which activate or inhibit SFK, respectively, or using the chemical inhibitor, PP2, with its inactive control, PP3. CCK (0.3, 100 nM) and TPA (1 μM) activated SFK and altered the activation of FAK proteins (PYK2, p125(FAK)), adaptor proteins (p130(CAS), paxillin), MAPK (p42/44, JNK, p38), Shc, PKC (PKD, MARCKS), Akt but not GSK3-β. Changes in SFK activity by using the three methods of altering SFK activity affected CCK/TPAs activation of SFK, PYK2, p125(FAK), p130(CAS), Shc, paxillin, Akt but not p42/44, JNK, p38, PKC (PKD, MARCKS) or GSK3-β. With chemical inhibition the active SFK inhibitor, PP2, but not the inactive control analogue, PP3, showed these effects. For all stimulated changes pre-incubation with both adenoviruses showed similar effects to chemical inhibition of SFK activity. In conclusion, using three different approaches to altering Src activity allowed us to define fully for the first time the roles of SFKs in acinar cell signaling. Our results show that in pancreatic acinar cells, SFKs play a much wider role than previously reported in activating a number of important cellular signaling cascades shown to be important in mediating both acinar cell physiological and pathophysiological responses.
Collapse
Affiliation(s)
- Bernardo Nuche-Berenguer
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1804, USA
| | - Paola Moreno
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1804, USA
| | - R. T. Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-1804, USA
| |
Collapse
|
17
|
Zhu R, Blazeski A, Poon E, Costa KD, Tung L, Boheler KR. Physical developmental cues for the maturation of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2014; 5:117. [PMID: 25688759 PMCID: PMC4396914 DOI: 10.1186/scrt507] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are the most promising source of cardiomyocytes (CMs) for experimental and clinical applications, but their use is largely limited by a structurally and functionally immature phenotype that most closely resembles embryonic or fetal heart cells. The application of physical stimuli to influence hPSC-CMs through mechanical and bioelectrical transduction offers a powerful strategy for promoting more developmentally mature CMs. Here we summarize the major events associated with in vivo heart maturation and structural development. We then review the developmental state of in vitro derived hPSC-CMs, while focusing on physical (electrical and mechanical) stimuli and contributory (metabolic and hypertrophic) factors that are actively involved in structural and functional adaptations of hPSC-CMs. Finally, we highlight areas for possible future investigation that should provide a better understanding of how physical stimuli may promote in vitro development and lead to mechanistic insights. Advances in the use of physical stimuli to promote developmental maturation will be required to overcome current limitations and significantly advance research of hPSC-CMs for cardiac disease modeling, in vitro drug screening, cardiotoxicity analysis and therapeutic applications.
Collapse
|
18
|
Honarpour N, Rose CM, Brumbaugh J, Anderson J, Graham RLJ, Sweredoski MJ, Hess S, Coon JJ, Deshaies RJ. F-box protein FBXL16 binds PP2A-B55α and regulates differentiation of embryonic stem cells along the FLK1+ lineage. Mol Cell Proteomics 2014; 13:780-91. [PMID: 24390425 PMCID: PMC3945908 DOI: 10.1074/mcp.m113.031765] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The programmed formation of specific tissues from embryonic stem cells is a major goal of regenerative medicine. To identify points of intervention in cardiac tissue formation, we performed an siRNA screen in murine embryonic stem cells to identify ubiquitin system genes that repress cardiovascular tissue formation. Our screen uncovered an F-box protein, Fbxl16, as a repressor of one of the earliest steps in the cardiogenic lineage: FLK1+ progenitor formation. Whereas F-box proteins typically form SCF ubiquitin ligases, shotgun mass spectrometry revealed that FBXL16 instead binds protein phosphatase 2A (PP2A) containing a B55 specificity subunit (PP2A(B55)). Phosphoproteomic analyses indicate that FBXL16 negatively regulates phosphorylation of the established PP2A(B55) substrate, vimentin. We suggest that FBXL16 negatively regulates the activity of B55α-PP2A to modulate the genesis of FLK1+ progenitor cells.
Collapse
|
19
|
Liao X, Lu S, Wu Y, Xu W, Zhuo Y, Peng Q, Li B, Zhang L, Wang Y. The effect of differentiation induction on FAK and Src activity in live HMSCs visualized by FRET. PLoS One 2013; 8:e72233. [PMID: 24015220 PMCID: PMC3754985 DOI: 10.1371/journal.pone.0072233] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 07/08/2013] [Indexed: 12/12/2022] Open
Abstract
FAK and Src signaling play important roles in cell differentiation, survival and migration. However, it remains unclear how FAK and Src activities are regulated at the initial stage of stem cell differentiation. We utilized fluorescence resonance energy transfer (FRET)-based FAK and Src biosensors to visualize these kinase activities at the plasma membrane of human mesenchymal stem cells (HMSCs) under the stimulation of osteogenic, myoblastic, or neural induction reagents. Our results indicate that the membrane FAK and Src activities are distinctively regulated by these differentiation induction reagents. FAK and Src activities were both up-regulated with positive feedback upon osteogenic induction, while myoblastic induction only activated Src, but not FAK. Neural induction, however, transiently activated FAK and subsequently Src, which triggered a negative feedback to partially inhibit FAK activity. These results unravel distinct regulation mechanisms of FAK and Src activities during HMSC fate decision, which should advance our understanding of stem cell differentiation in tissue engineering.
Collapse
Affiliation(s)
- Xiaoling Liao
- Biomaterials and Live Cell Imaging Institute, Chongqing University of Science and technology, Chongqing, People's Republic of China
- Beckman Institute for Advanced Science and Technology, Center for Biophysics and Computational Biology, Department of Integrative and Molecular Physiology, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Shaoying Lu
- Department of Bioengineering, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Bioengineering, University of California San Diego, San Diego, California, United States of America
| | - Yiqian Wu
- Biomedical Engineering Programme, Department of Electronic Engineering, Chinese University of Hong Kong, Shatin, NT, Hong Kong, People's Republic of China
| | - Wenfeng Xu
- Biomaterials and Live Cell Imaging Institute, Chongqing University of Science and technology, Chongqing, People's Republic of China
| | - Yue Zhuo
- Department of Bioengineering, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Qin Peng
- Department of Bioengineering, University of California San Diego, San Diego, California, United States of America
| | - Bo Li
- Biomaterials and Live Cell Imaging Institute, Chongqing University of Science and technology, Chongqing, People's Republic of China
| | - Ling Zhang
- Biomaterials and Live Cell Imaging Institute, Chongqing University of Science and technology, Chongqing, People's Republic of China
| | - Yingxiao Wang
- Department of Bioengineering, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
- Beckman Institute for Advanced Science and Technology, Center for Biophysics and Computational Biology, Department of Integrative and Molecular Physiology, Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Bioengineering, University of California San Diego, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
20
|
Parameswaran S, Kumar S, Verma RS, Sharma RK. Cardiomyocyte culture - an update on the in vitro cardiovascular model and future challenges. Can J Physiol Pharmacol 2013; 91:985-98. [PMID: 24289068 DOI: 10.1139/cjpp-2013-0161] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The success of any work with isolated cardiomyocytes depends on the reproducibility of cell isolation, because the cells do not divide. To date, there is no suitable in vitro model to study human adult cardiac cell biology. Although embryonic stem cells and induced pluripotent stem cells are able to differentiate into cardiomyocytes in vitro, the efficiency of this process is low. Isolation and expansion of human cardiomyocyte progenitor cells from cardiac surgical waste or, alternatively, from fetal heart tissue is another option. However, to overcome various issues related to human tissue usage, especially ethical concerns, researchers use large- and small-animal models to study cardiac pathophysiology. A simple model to study the changes at the cellular level is cultures of cardiomyocytes. Although primary murine cardiomyocyte cultures have their own advantages and drawbacks, alternative strategies have been developed in the last two decades to minimise animal usage and interspecies differences. This review discusses the use of freshly isolated murine cardiomyocytes and cardiomyocyte alternatives for use in cardiac disease models and other related studies.
Collapse
Affiliation(s)
- Sreejit Parameswaran
- a Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | | | | | | |
Collapse
|
21
|
Gong J, Gu HY, Wang X, Liang Y, Sun T, Liu PJ, Wang Y, Yan JC, Jiao ZJ. SRC kinase family inhibitor PP2 promotes DMSO-induced cardiac differentiation of P19 cells and inhibits proliferation. Int J Cardiol 2013; 167:1400-5. [DOI: 10.1016/j.ijcard.2012.04.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 02/21/2012] [Accepted: 04/08/2012] [Indexed: 10/28/2022]
|
22
|
Lee JS, Ha L, Kwon IK, Lim JY. The role of focal adhesion kinase in BMP4 induction of mesenchymal stem cell adipogenesis. Biochem Biophys Res Commun 2013; 435:696-701. [DOI: 10.1016/j.bbrc.2013.05.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/10/2013] [Indexed: 10/26/2022]
|
23
|
Schukur L, Zorlutuna P, Cha JM, Bae H, Khademhosseini A. Directed differentiation of size-controlled embryoid bodies towards endothelial and cardiac lineages in RGD-modified poly(ethylene glycol) hydrogels. Adv Healthc Mater 2013; 2:195-205. [PMID: 23193099 PMCID: PMC3635117 DOI: 10.1002/adhm.201200194] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Revised: 07/24/2012] [Indexed: 12/26/2022]
Abstract
Recent advances in stem cell research have demonstrated the importance of microenvironmental cues in directing stem cell fate towards specific cell lineages. For instance, the size of the embryoid body (EB) was shown to play a role in stem cell differentiation. Other studies have used cell adhesive RGD peptides to direct stem cell fate towards endothelial cells. In this study, materials and cell-based approaches are combined by using microwell arrays to produce size-controlled EBs and encapsulating the resulting aggregates in high molecular weight PEG-4 arm acrylate with and without conjugated RGD to study their effect on stem cell differentiation in a 3D microenvironment. Increasing EB size is observed along with a decrease in the total number of EBs in pristine PEG hydrogel, regardless of the initial EB size. In correlation with this aggregation, EBs in PEG show enhanced cardiogenic differentiation compared to RGD-PEG hydrogel. Both aggregation and cardiogenic differentiation are significantly reduced when RGD peptides are introduced to the microenvironment, while endothelial cell differentiation is accelerated by 3 to 5 days, depending on the EB size, and doubled over the course of cell culture for both EB sizes. Presented results indicate that RGD sequence has a dominant effect in driving endothelial cell differentiation in size-controlled EBs, while pristine multi-arm, high molecular weight PEG can induce cardiogenic differentiation, possibly through EB aggregation. The photopatternable nature of the hydrogel used in this study enabled patterning of such domains devoid or abundant of cell attachment sequences. Therefore, these hydrogels can potentially be used for spatially patterned embryonic stem cell differentiation, which may be beneficial for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Lina Schukur
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, USA, 65 Landsdowne Street Cambridge, MA 02139, USA
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52074, Germany
| | - Pinar Zorlutuna
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, USA, 65 Landsdowne Street Cambridge, MA 02139, USA
| | - Jae Min Cha
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, USA, 65 Landsdowne Street Cambridge, MA 02139, USA
| | - Hojae Bae
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, USA, 65 Landsdowne Street Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 02139, USA, 65 Landsdowne Street Cambridge, MA 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 02115, USA
| |
Collapse
|
24
|
Pentassuglia L, Sawyer DB. ErbB/integrin signaling interactions in regulation of myocardial cell-cell and cell-matrix interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:909-16. [PMID: 23261977 DOI: 10.1016/j.bbamcr.2012.12.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 12/02/2012] [Accepted: 12/03/2012] [Indexed: 12/17/2022]
Abstract
Neuregulin (Nrg)/ErbB and integrin signaling pathways are critical for the normal function of the embryonic and adult heart. Both systems activate several downstream signaling pathways, with different physiological outputs: cell survival, fibrosis, excitation-contraction coupling, myofilament structure, cell-cell and cell-matrix interaction. Activation of ErbB2 by Nrg1β in cardiomycytes or its overexpression in cancer cells induces phosphorylation of FAK (Focal Adhesion Kinase) at specific sites with modulation of survival, invasion and cell-cell contacts. FAK is also a critical mediator of integrin receptors, converting extracellular matrix alterations into intracellular signaling. Systemic FAK deletion is lethal and is associated with left ventricular non-compaction whereas cardiac restriction in adult hearts is well tolerated. Nevertheless, these hearts are more susceptible to stress conditions like trans-aortic constriction, hypertrophy, and ischemic injury. As FAK is both downstream and specifically activated by integrins and Nrg-1β, here we will explore the role of FAK in the heart as a protective factor and as possible mediator of the crosstalk between the ErbB and Integrin receptors. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.
Collapse
|
25
|
Kim DH, Kshitiz, Smith RR, Kim P, Ahn EH, Kim HN, Marbán E, Suh KY, Levchenko A. Nanopatterned cardiac cell patches promote stem cell niche formation and myocardial regeneration. Integr Biol (Camb) 2012; 4:1019-33. [PMID: 22890784 DOI: 10.1039/c2ib20067h] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stem cell-based methods for myocardial regeneration suffer from considerable cell attrition. Artificial matrices reproducing mechanical and structural properties of the native tissue may facilitate survival, retention and functional integration of adult stem or progenitor cells, by conditioning the cells prior to, and during, transplantation. Here we combined autologous cardiosphere-derived cells (CDCs) with nanotopographically defined hydrogels mimicking the native myocardial matrix, to form in vitro cardiac stem cell niches, and control cell function and fate. These platforms were used to produce cardiac patches that could be transplanted at the site of infarct. In culture, highly anisotropic, but not more randomized nanotopographic, control augmented cell adhesion, migration, and proliferation. It also dramatically enhanced early, and, in the presence of mature cardiomyocytes, late cardiomyogenesis. Nanotopography sensing and transcriptional response was mediated via p190RhoGAP. In a rat infarction model, engraftment of nanofabricated scaffolds with CDCs enhanced retention and growth of transplanted cells, and their integration with the host tissue. The infarcted ventricle wall increased in thickness, with higher cell viability and better collagen organization. These results suggest that nanostructured polymeric materials that closely mimic the extracellular matrix structure on which cardiac cells reside in vivo can be both very effective tools in investigating the mechanisms of cardiac differentiation and the basis for cardiac tissue engineering, thus facilitating stem cell-based therapy in the heart.
Collapse
Affiliation(s)
- Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Liao X, Lu S, Zhuo Y, Winter C, Xu W, Wang Y. Visualization of Src and FAK activity during the differentiation process from HMSCs to osteoblasts. PLoS One 2012; 7:e42709. [PMID: 22900044 PMCID: PMC3416797 DOI: 10.1371/journal.pone.0042709] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 07/11/2012] [Indexed: 12/14/2022] Open
Abstract
Non-receptor protein kinases FAK and Src play crucial roles in regulating cellular adhesions, growth, migration and differentiation. However, it remains unclear how the activity of FAK and Src is regulated during the differentiation process from mesenchymal stem cells (MSCs) to bone cells. In this study, we used genetically encoded FAK and Src biosensors based on fluorescence resonance energy transfer (FRET) to monitor the FAK and Src activity in live cells during the differentiation process. The results revealed that the FAK activity increased after the induction of differentiation, which peaked around 20–27 days after induction. Meanwhile, the Src activity decreased continuously for 27 days after induction. Therefore, the results showed significant and differential changes of FAK and Src activity upon induction. This opposite trend between FAK and Src activation suggests novel and un-coupled Src/FAK functions during the osteoblastic differentiation process. These results should provide important information for the biochemical signals during the differentiation process of stem cells toward bone cells, which will advance our understanding of bone repair and tissue engineering.
Collapse
Affiliation(s)
- Xiaoling Liao
- Biomaterials and Live Cell Imaging Institute, School of Metallurgy and Materials Engineering, Chongqing University of Science and technology, Chongqing, People’s Republic of China
- Beckman Institute for Advanced Science and Technology, Center for Biophysics and Computational Biology, Institute for Genomic Biology, Department of Integrative and Molecular Physiology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Shaoying Lu
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yue Zhuo
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Christina Winter
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
| | - Wenfeng Xu
- Biomaterials and Live Cell Imaging Institute, School of Metallurgy and Materials Engineering, Chongqing University of Science and technology, Chongqing, People’s Republic of China
| | - Yingxiao Wang
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
- Beckman Institute for Advanced Science and Technology, Center for Biophysics and Computational Biology, Institute for Genomic Biology, Department of Integrative and Molecular Physiology, University of Illinois, Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
27
|
Li Q, Ramírez-Bergeron DL, Dunwoodie SL, Yang YC. Cited2 gene controls pluripotency and cardiomyocyte differentiation of murine embryonic stem cells through Oct4 gene. J Biol Chem 2012; 287:29088-100. [PMID: 22761414 DOI: 10.1074/jbc.m112.378034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cited2 (CBP/p300-interacting transactivator with glutamic acid (E)/aspartic acid (D)-rich tail 2) is a transcriptional modulator critical for the development of multiple organs. Although many Cited2-mediated phenotypes and molecular events have been well characterized using in vivo genetic murine models, Cited2-directed cell fate decision in embryonic stem cells (ESCs) remains elusive. In this study, we examined the role of Cited2 in the maintenance of stemness and pluripotency of murine ESCs by a gene-targeting approach. Cited2 knock-out (Cited2(Δ/-), KO) ESCs display defective differentiation. Loss of Cited2 in differentiating ESCs results in delayed silencing of the genes involved in the maintenance of pluripotency and self-renewal of stem cells (Oct4, Klf4, Sox2, and c-Myc) and the disturbance in cardiomyocyte, hematopoietic, and neuronal differentiation. In addition, Cited2 KO ESCs experience a delayed induction of cardiomyocyte differentiation-associated proteins, NFAT3 (along with the reduced expression of NFAT3 target genes, Nkx2.5 and β-MHC), N-cadherin, and smooth muscle actin. CITED2 is recruited to the Oct4 promoter to regulate its expression during early ESC differentiation. This is the first demonstration that Cited2 controls ESC pluripotency and differentiation via direct regulation of Oct4 gene expression.
Collapse
Affiliation(s)
- Qiang Li
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
28
|
Combinatorial polymer electrospun matrices promote physiologically-relevant cardiomyogenic stem cell differentiation. PLoS One 2011; 6:e28935. [PMID: 22216144 PMCID: PMC3246450 DOI: 10.1371/journal.pone.0028935] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 11/17/2011] [Indexed: 01/08/2023] Open
Abstract
Myocardial infarction results in extensive cardiomyocyte death which can lead to fatal arrhythmias or congestive heart failure. Delivery of stem cells to repopulate damaged cardiac tissue may be an attractive and innovative solution for repairing the damaged heart. Instructive polymer scaffolds with a wide range of properties have been used extensively to direct the differentiation of stem cells. In this study, we have optimized the chemical and mechanical properties of an electrospun polymer mesh for directed differentiation of embryonic stem cells (ESCs) towards a cardiomyogenic lineage. A combinatorial polymer library was prepared by copolymerizing three distinct subunits at varying molar ratios to tune the physicochemical properties of the resulting polymer: hydrophilic polyethylene glycol (PEG), hydrophobic poly(ε-caprolactone) (PCL), and negatively-charged, carboxylated PCL (CPCL). Murine ESCs were cultured on electrospun polymeric scaffolds and their differentiation to cardiomyocytes was assessed through measurements of viability, intracellular reactive oxygen species (ROS), α-myosin heavy chain expression (α-MHC), and intracellular Ca(2+) signaling dynamics. Interestingly, ESCs on the most compliant substrate, 4%PEG-86%PCL-10%CPCL, exhibited the highest α-MHC expression as well as the most mature Ca(2+) signaling dynamics. To investigate the role of scaffold modulus in ESC differentiation, the scaffold fiber density was reduced by altering the electrospinning parameters. The reduced modulus was found to enhance α-MHC gene expression, and promote maturation of myocyte Ca(2+) handling. These data indicate that ESC-derived cardiomyocyte differentiation and maturation can be promoted by tuning the mechanical and chemical properties of polymer scaffold via copolymerization and electrospinning techniques.
Collapse
|
29
|
Hiradate Y, Ohtake J, Hoshino Y, Tanemura K, Sato E. Adrenomedullin: a possible regulator of germinal vesicle breakdown. Biochem Biophys Res Commun 2011; 415:691-5. [PMID: 22079286 DOI: 10.1016/j.bbrc.2011.10.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 10/28/2011] [Indexed: 10/15/2022]
Abstract
Adrenomedullin (ADM) is a multifunctional hormone that regulates processes as diverse as blood pressure and cell growth. Although expressed in the ovary, the role of ADM in this organ is not clear. In the present study, we found the expression of ADM receptor and receptor activity-modifying proteins in mouse cumulus cells but not in the oocytes. We report that germinal vesicle breakdown (GVBD), which is required for oocyte maturation, is not inhibited by ADM alone. However, ADM in the presence of the nitric oxide donor sodium nitroprusside (SNP) significantly inhibited GVBD. Furthermore, the ADM- and SNP-dependent inhibition of GVBD was abrogated by Akt blockade. Additionally, Akt expression and phosphorylation was exhibited by ADM, suggesting that Akt signaling upstream in cumulus cells is responsible. Additionally, immunohistochemical analysis revealed that ADM was localized in the granulosa cells of developed follicles, implying the possibility that ADM physiologically affects oocyte maturation in vivo. Our results provide the evidence that ADM can act as a GVBD regulator.
Collapse
Affiliation(s)
- Yuuki Hiradate
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi Aobaku, Sendai 981-8555, Japan.
| | | | | | | | | |
Collapse
|
30
|
Afrikanova I, Yebra M, Simpkinson M, Xu Y, Hayek A, Montgomery A. Inhibitors of Src and focal adhesion kinase promote endocrine specification: impact on the derivation of β-cells from human pluripotent stem cells. J Biol Chem 2011; 286:36042-36052. [PMID: 21852242 DOI: 10.1074/jbc.m111.290825] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Stepwise approaches for the derivation of β-cells from human embryonic stem cells have been described. However, low levels of endocrine specification limit the final yield of insulin-producing β-cells. In this study, we show that the pyrrolo-pyrimidine Src family kinase (SFK) inhibitor PP2 effectively promotes the endocrine specification of human embryonic stem cell derivatives based on its capacity to induce the expression of proendocrine transcription factors (NGN3, NEUROD1, NKX2.2, and PAX4) and to significantly increase the final yield of insulin-positive cells. We further demonstrate that PP2 inhibits the activation of focal adhesion kinase (FAK), and selective inhibition of this kinase is also sufficient to induce early endocrine commitment based on increased expression of NGN3, NEUROD1, and NKX2.2. Additional studies using dominant negative constructs and isolated human fetal pancreata suggest that c-Src is at least partially responsible for inhibiting early endocrine specification. Mechanistically, we propose that inhibition of SFK/FAK signaling can promote endocrine specification by limiting activation of the TGFβR/Smad2/3 pathway. Moreover, we show that inhibition of SFK/FAK signaling suppresses cell growth, increases the expression of the β-cell-associated cyclin-dependent kinase inhibitor p57kip2, and simultaneously suppresses the expression of Id1 and Id2. This study has important implications for the derivation of β-cells for the cell-based therapy of diabetes and sheds new light on the signaling events that regulate early endocrine specification.
Collapse
Affiliation(s)
- Ivka Afrikanova
- Department of Pediatrics, University of California San Diego, San Diego, California 92121; Pediatric Diabetes Research Center, University of California San Diego, San Diego, California 92121
| | - Mayra Yebra
- Department of Pediatrics, University of California San Diego, San Diego, California 92121; Pediatric Diabetes Research Center, University of California San Diego, San Diego, California 92121
| | - Megan Simpkinson
- Department of Pediatrics, University of California San Diego, San Diego, California 92121; Pediatric Diabetes Research Center, University of California San Diego, San Diego, California 92121
| | - Yang Xu
- Division of Biological Science, University of California San Diego, San Diego, California 92121
| | - Alberto Hayek
- Department of Pediatrics, University of California San Diego, San Diego, California 92121; Pediatric Diabetes Research Center, University of California San Diego, San Diego, California 92121
| | - Anthony Montgomery
- Department of Pediatrics, University of California San Diego, San Diego, California 92121; Pediatric Diabetes Research Center, University of California San Diego, San Diego, California 92121.
| |
Collapse
|
31
|
Schenke-Layland K, Nsair A, Van Handel B, Angelis E, Gluck JM, Votteler M, Goldhaber JI, Mikkola HK, Kahn M, Maclellan WR. Recapitulation of the embryonic cardiovascular progenitor cell niche. Biomaterials 2011; 32:2748-56. [PMID: 21257198 PMCID: PMC3414535 DOI: 10.1016/j.biomaterials.2010.12.046] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 12/28/2010] [Indexed: 01/16/2023]
Abstract
Stem or progenitor cell populations are often established in unique niche microenvironments that regulate cell fate decisions. Although niches have been shown to be critical for the normal development of several tissues, their role in the cardiovascular system is poorly understood. In this study, we characterized the cardiovascular progenitor cell (CPC) niche in developing human and mouse hearts, identifying signaling pathways and extracellular matrix (ECM) proteins that are crucial for CPC maintenance and expansion. We demonstrate that collagen IV (ColIV) and β-catenin-dependent signaling are essential for maintaining and expanding undifferentiated CPCs. Since niches are three-dimensional (3D) structures, we investigated the impact of a 3D microenvironment that mimics the in vivo niche ECM. Employing electrospinning technologies, 3D in vitro niche substrates were bioengineered to serve as culture inserts. The three-dimensionality of these structures increased mouse embryonic stem cell differentiation into CPCs when compared to 2D control cultures, which was further enhanced by incorporation of ColIV into the substrates. Inhibiting p300-dependent β-catenin signals with the small molecule IQ1 facilitated further expansion of CPCs. Our study represents an innovative approach to bioengineer cardiac niches that can serve as unique 3D in vitro systems to facilitate CPC expansion and study CPC biology.
Collapse
Affiliation(s)
- Katja Schenke-Layland
- Department of Cell and Tissue Engineering, Fraunhofer IGB, 70569 Stuttgart, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Endothelial focal adhesion kinase mediates cancer cell homing to discrete regions of the lungs via E-selectin up-regulation. Proc Natl Acad Sci U S A 2011; 108:3725-30. [PMID: 21321210 DOI: 10.1073/pnas.1100446108] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Primary tumors secrete factors that alter the microenvironment of distant organs, rendering those organs as fertile soil for subsequent metastatic cancer cell colonization. Although the lungs are exposed to these factors ubiquitously, lung metastases usually develop as a series of discrete lesions. The underlining molecular mechanisms of the formation of these discrete lesions are not understood. Here we show that primary tumors induce formation of discrete foci of vascular hyperpermeability in premetastatic lungs. This is mediated by endothelial cell-focal adhesion kinase (FAK), which up-regulates E-selectin, leading to preferential homing of metastatic cancer cells to these foci. Suppression of endothelial-FAK or E-selectin activity attenuates the number of cancer cells homing to these foci. Thus, localized activation of endothelial FAK and E-selectin in the lung vasculature mediates the initial homing of metastatic cancer cells to specific foci in the lungs.
Collapse
|
33
|
Abstract
Embryonic stem (ES) cells and their differentiated progeny offer tremendous potential for regenerative medicine, even in the field of drug discovery. There is an urgent need for clinically relevant assays that make use of ES cells because of their rich biological utility. Attention has been focused on small molecules that allow the precise manipulation of cells in vitro, which could allow researchers to obtain homogeneous cell types for cell-based therapies and discover drugs for stimulating the regeneration of endogenous cells. Such therapeutics can act on target cells or their niches in vivo to promote cell survival, proliferation, differentiation, and homing. In the present paper, we reviewed the use of ES cell models for high-throughput/content drug screening and toxicity assessment. In addition, we examined the role of stem cells in large pharmaceutical companies' R&D and discussed a novel subject, nicheology, in stem cell-related research fields.
Collapse
|
34
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemical Control of Stem Cell Fate and Developmental Potential. Angew Chem Int Ed Engl 2010; 50:200-42. [DOI: 10.1002/anie.201004284] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
35
|
Lyssiotis CA, Lairson LL, Boitano AE, Wurdak H, Zhu S, Schultz PG. Chemische Kontrolle des Schicksals und Entwicklungspotenzials von Stammzellen. Angew Chem Int Ed Engl 2010. [DOI: 10.1002/ange.201004284] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Costas A. Lyssiotis
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Luke L. Lairson
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Anthony E. Boitano
- The Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121 (USA)
| | - Heiko Wurdak
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Shoutian Zhu
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| | - Peter G. Schultz
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037 (USA), Fax: (+1) 858‐784‐9440
| |
Collapse
|
36
|
Chen M, Lin YQ, Xie SL, Wu HF, Wang JF. Enrichment of cardiac differentiation of mouse embryonic stem cells by optimizing the hanging drop method. Biotechnol Lett 2010; 33:853-8. [PMID: 21165673 DOI: 10.1007/s10529-010-0494-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 12/01/2010] [Indexed: 11/28/2022]
Abstract
Hanging drop (HD) culture is used to induce differentiation of embryonic stem cells (ESCs) into other cell types including cardiomyocytes. However, the factors affecting cardiac differentiation of ESCs with this method remain incompletely understood. We have investigated the effects of the starting number of ESCs in embryoid bodies (EBs) and the time of EB adherence to gelatin-coated plates on cardiac differentiation: cardiac differentiation was increased in the EBs by a larger number of ESCs and was decreased by plating EBs at day 4 or earlier. These two factors can thus be optimized to enrich the cardiac differentiation in ESCs using the HD method.
Collapse
Affiliation(s)
- Ming Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
37
|
Huang NF, Patlolla B, Abilez O, Sharma H, Rajadas J, Beygui RE, Zarins CK, Cooke JP. A matrix micropatterning platform for cell localization and stem cell fate determination. Acta Biomater 2010; 6:4614-21. [PMID: 20601236 PMCID: PMC2957527 DOI: 10.1016/j.actbio.2010.06.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 06/26/2010] [Accepted: 06/28/2010] [Indexed: 11/28/2022]
Abstract
To study the role of cell-extracellular matrix (ECM) interactions, microscale approaches provide the potential to perform high throughput assessment of the effect of the ECM microenvironment on cellular function and phenotype. Using a microscale direct writing (MDW) technique, we characterized the generation of multicomponent ECM microarrays for cellular micropatterning, localization and stem cell fate determination. ECMs and other biomolecules of various geometries and sizes were printed onto epoxide-modified glass substrates to evaluate cell attachment by human endothelial cells. The endothelial cells displayed strong preferential attachment to the ECM patterned regions and aligned their cytoskeleton along the direction of the micropatterns. We next generated ECM microarrays that contained one or more ECM components (namely gelatin, collagen IV and fibronectin) and then cultured murine embryonic stem cell (ESCs) on the microarrays. The ESCs selectively attached to the micropatterned features and expressed markers associated with a pluripotent phenotype, such as E-cadherin and alkaline phosphatase, when maintained in growth medium containing leukemia inhibitory factor. In the presence of the soluble factors retinoic acid and bone morphogenetic protein-4 the ESCs differentiated towards the ectodermal lineage on the ECM microarray with differential ECM effects. The ESCs cultured on gelatin showed significantly higher levels of pan cytokeratin expression, when compared with cells cultured on collagen IV or fibronectin, suggesting that gelatin preferentially promotes ectodermal differentiation. In summary, our results demonstrate that MDW is a versatile approach to print ECMs of diverse geometries and compositions onto surfaces, and it is amenable to the generation of multicomponent ECM microarrays for stem cell fate determination.
Collapse
Affiliation(s)
- Ngan F. Huang
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA. Fax: 650-725-1599; Tel: 650-723-0899
| | - Bhagat Patlolla
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA. Fax: 650- 725-3846; Tel: 650-724-0831
| | - Oscar Abilez
- Department of Surgery, Stanford University, Stanford, CA, USA. Fax: 650-498-6044; Tel: 650-725-7830
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Himanshu Sharma
- Department of Surgery, Stanford University, Stanford, CA, USA. Fax: 650-498-6044; Tel: 650-725-7830
| | - Jaykumar Rajadas
- Biomaterial and Advanced Drug Delivery Center, Stanford University, Stanford, CA, USA
| | - Ramin E. Beygui
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA. Fax: 650- 725-3846; Tel: 650-724-0831
| | - Christopher K. Zarins
- Department of Surgery, Stanford University, Stanford, CA, USA. Fax: 650-498-6044; Tel: 650-725-7830
| | - John P. Cooke
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA, USA. Fax: 650-725-1599; Tel: 650-723-0899
| |
Collapse
|
38
|
Zhang L, Wang D, Jiang W, Edwards D, Qiu W, Barroilhet LM, Rho JH, Jin L, Seethappan V, Vitonis A, Wang J, Mok SC, Crum C, Cramer DW, Ye B. Activated networking of platelet activating factor receptor and FAK/STAT1 induces malignant potential in BRCA1-mutant at-risk ovarian epithelium. Reprod Biol Endocrinol 2010; 8:74. [PMID: 20576130 PMCID: PMC2903602 DOI: 10.1186/1477-7827-8-74] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 06/24/2010] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVES It is essential to understand the molecular basis of ovarian cancer etiology and tumor development to provide more effective preventive and therapeutic approaches to reduce mortality. Particularly, the molecular targets and pathways involved in early malignant transformation are still not clear. Pro-inflammatory lipids and pathways have been reported to play significant roles in ovarian cancer progression and metastasis. The major objective of this study was to explore and determine whether platelet activating factor (PAF) and receptor associated networking pathways might significantly induce malignant potential in BRCA1-mutant at-risk epithelial cells. METHODS BRCA1-mutant ovarian epithelial cell lines including (HOSE-636, HOSE-642), BRCA1-mutant ovarian cancer cell (UWB1.289), wild type normal ovarian epithelial cell (HOSE-E6E7) and cancerous cell line (OVCA429), and the non-malignant BRCA1-mutant distal fallopian tube (fimbria) tissue specimens were used in this study. Mutation analysis, kinase microarray, western blot, immune staining, co-immune precipitation, cell cycle, apoptosis, proliferation and bioinformatic pathway analysis were applied. RESULTS We found that PAF, as a potent pro-inflammatory mediator, induced significant anti-apoptotic effect in BRCA1-mutant ovarian surface epithelial cells, but not in wild type HOSE cells. With kinase microarray technology and the specific immune approaches, we found that phosphor-STAT1 was activated by 100 nM PAF treatment only in BRCA1-mutant associated at-risk ovarian epithelial cells and ovarian cancer cells, but not in BRCA1-wild type normal (HOSE-E6E7) or malignant (OVCA429) ovarian epithelial cells. Co-immune precipitation revealed that elevated PAFR expression is associated with protein-protein interactions of PAFR-FAK and FAK-STAT1 in BRCA1-mutant ovarian epithelial cells, but not in the wild-type control cells. CONCLUSION Previous studies showed that potent inflammatory lipid mediators such as PAF and its receptor (PAFR) significantly contribute to cancer progression and metastasis. Our findings suggest that these potent inflammatory lipids and receptor pathways are significantly involved in the early malignant transformation through PAFR-FAK-STAT1 networking and to block apoptosis pathway in BRCA1 dysfunctional at-risk ovarian epithelium.
Collapse
Affiliation(s)
- Lifang Zhang
- Obstetrics and Gynecology Department, Peking University People's Hospital, Beijing, China
| | - Dan Wang
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Wei Jiang
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fang Xie Road, Shanghai 200011, China
| | - Dale Edwards
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Weiliang Qiu
- Channing Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Lisa M Barroilhet
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jung-hyun Rho
- Channing Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Lianjin Jin
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Vanitha Seethappan
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Allison Vitonis
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jianliu Wang
- Obstetrics and Gynecology Department, Peking University People's Hospital, Beijing, China
| | - Samuel C Mok
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Christopher Crum
- Department Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel W Cramer
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| | - Bin Ye
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
39
|
Differentiation of mesodermal cells from pluripotent stem cells. Int J Hematol 2010; 91:373-83. [PMID: 20224874 DOI: 10.1007/s12185-010-0518-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 01/15/2010] [Accepted: 01/21/2010] [Indexed: 01/12/2023]
Abstract
The pluripotency of embryonic stem cells has been well demonstrated by a vast variety of studies showing the induction of differentiation into desired cell types that have the potential to be used not only in basic studies but also in medical applications. The induction of mesodermal cells, especially blood cells, from embryonic stem cells is notable from the point of view of transplantation, and the methods for this induction have improved over the last few years, with more defined culture conditions in place. Concurrently, the generation of induced pluripotent stem cells from somatic cells opens the possibility of autologous transplantation. In fact, there are a growing number of reports demonstrating that several mesodermal cells can be differentiated from induced pluripotent stem cells using the same methods used for embryonic stem cells. This review summarizes recent advances in the differentiation of mesodermal cells from embryonic stem cells and induced pluripotent stem cells.
Collapse
|
40
|
Papp S, Dziak E, Opas M. Embryonic stem cell-derived cardiomyogenesis: a novel role for calreticulin as a regulator. Stem Cells 2009; 27:1507-15. [PMID: 19544459 DOI: 10.1002/stem.85] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A role for calreticulin, an endoplasmic reticulum (ER)-resident, Ca(2+)-binding chaperone, has recently emerged in the context of cardiomyogenesis. We previously proposed calreticulin to be a novel cardiac fetal gene, because calreticulin knockout causes embryonic lethality in mice as a result of cardiac defects, it is transiently activated during heart development, and heart-targeted overexpression of constitutively active calcineurin in calreticulin-null mice rescues the lethal phenotype. Calreticulin affects Ca(2+) homeostasis and expression of adhesion-related genes. Using cardiomyocytes derived from both calreticulin-null and wild-type embryonic stem (ES) cells, we show here that cardiomyogenesis from calreticulin-null ES cells is accelerated but deregulated, such that the myofibrils of calreticulin-null cardiomyocytes become disorganized and disintegrate with time in culture. We have previously shown that the disorganization of the actin cytoskeleton in calreticulin-null cells may be explained, at least in part, by the downregulation of adhesion proteins, implying that calreticulin ablation causes adhesion-related defects. Here, upon examination of adhesion proteins, we found that vinculin is downregulated in calreticulin-null cardiomyocytes. We also found c-Src activity to be higher in calreticulin-null cardiomyocytes than in wild-type cardiomyocytes, and c-Src activity is affected by both calreticulin and [Ca(2+)]. Finally, we show that calreticulin and calsequestrin, the major Ca(2+) storage proteins of the ER and sarcoplasmic reticulum, respectively, exhibit alternate distributions. This suggests that calreticulin may have a housekeeping role to play in mature cardiomyocytes as well as during cardiomyogenesis. We propose here that calreticulin, an ER Ca(2+) storage protein, is a crucial regulator of cardiomyogenesis whose presence is required for controlled cardiomyocyte development from ES cells.
Collapse
Affiliation(s)
- Sylvia Papp
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
41
|
Zuk PA. The intracellular distribution of the ES cell totipotent markers OCT4 and Sox2 in adult stem cells differs dramatically according to commercial antibody used. J Cell Biochem 2009; 106:867-77. [PMID: 19199344 DOI: 10.1002/jcb.22054] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
To characterize ES cells, researchers have at their disposal a list of pluripotent markers, such as OCT4. In their quest to determine if adult stem cell populations, such as MSCs and ASCs, are pluripotent, several groups have begun to report the expression of these markers in these cells. Consistent with this, human ASCs (hASCs) are shown in this study to express a plethora of ES pluripotent markers at the gene and protein level, including OCT4, Sox2, and Nanog. When intracellular distribution is examined in hASCs, both OCT4 and Sox2 are expressed within the nuclei of hASCs, consistent with their expression patterns in ES cells. However, a significant amount of expression can be noted within the hASC cytoplasm and a complete absence of nuclear expression is observed for Nanog. Recent descriptions of OCT4 transcript variants may explain the cytoplasmic expression of OCT4 in hASCs and consistent with this, hASCs do express both the OCT4A and 4B transcript variants at the gene level. However, discrepancies arise when these three pluripotent markers are studied at the protein level. Specifically, distinct differences in intracellular expression patterns were noted for OCT4, Sox2, and Nanog from commercial antibody to commercial antibody. These antibody discrepancies persisted when hMSCs and rat ASCs and MSCs were examined. Therefore, confirming the expression of OCT4, Sox2, and Nanog in adult stem cells with today's commercial antibodies must be carefully considered before the designation of pluripotent can be granted.
Collapse
Affiliation(s)
- Patricia A Zuk
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| |
Collapse
|
42
|
Li H, Xing X, Ding G, Li Q, Wang C, Xie L, Zeng R, Li Y. SysPTM: a systematic resource for proteomic research on post-translational modifications. Mol Cell Proteomics 2009; 8:1839-49. [PMID: 19366988 DOI: 10.1074/mcp.m900030-mcp200] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
With the rapid expansion of protein post-translational modification (PTM) research based on large-scale proteomic work, there is an increasing demand for a suitable repository to analyze PTM data. Here we present a curated, web-accessible PTM data base, SysPTM. SysPTM provides a systematic and sophisticated platform for proteomic PTM research equipped not only with a knowledge base of manually curated multi-type modification data but also with four fully developed, in-depth data mining tools. Currently, SysPTM contains data detailing 117,349 experimentally determined PTM sites on 33,421 proteins involving nearly 50 PTM types, curated from public resources including five data bases and four web servers and more than one hundred peer-reviewed mass spectrometry papers. Protein annotations including Pfam domains, KEGG pathways, GO functional classification, and ortholog groups are integrated into the data base. Four online tools have been developed and incorporated, including PTMBlast, to compare a user's PTM dataset with PTM data in SysPTM; PTMPathway, to map PTM proteins to KEGG pathways; PTMPhylog, to discover potentially conserved PTM sites; and PTMCluster, to find clusters of multi-site modifications. The workflow of SysPTM was demonstrated by analyzing an in-house phosphorylation dataset identified by MS/MS. It is shown that in SysPTM, the role of single-type and multi-type modifications can be systematically investigated in a full biological context. SysPTM could be an important contribution to modificomics research. SysPTM is freely available online at www.sysbio.ac.cn/SysPTM.
Collapse
Affiliation(s)
- Hong Li
- Key Lab of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Franchini K, Clemente C, Marin T. Focal adhesion kinase signaling in cardiac hypertrophy and failure. Braz J Med Biol Res 2009; 42:44-52. [DOI: 10.1590/s0100-879x2009000100008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Accepted: 12/11/2008] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - T.M. Marin
- Universidade Estadual de Campinas, Brasil
| |
Collapse
|
44
|
Sayers RL, Sundberg-Smith LJ, Rojas M, Hayasaka H, Parsons JT, Mack CP, Taylor JM. FRNK expression promotes smooth muscle cell maturation during vascular development and after vascular injury. Arterioscler Thromb Vasc Biol 2008; 28:2115-22. [PMID: 18787183 PMCID: PMC2785047 DOI: 10.1161/atvbaha.108.175455] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Smooth muscle cell (SMC) differentiation is a dynamic process that must be tightly regulated for proper vascular development and to control the onset of vascular disease. Our laboratory previously reported that a specific focal adhesion kinase (FAK) inhibitor termed FRNK (FAK Related Non-Kinase) is selectively expressed in large arterioles when SMCs are transitioning from a synthetic to contractile phenotype and that FRNK inhibits FAK-dependent SMC proliferation and migration. Herein, we sought to determine whether FRNK expression modulates SMC phenotypes in vivo. METHODS AND RESULTS We present evidence that FRNK(-/-) mice exhibit attenuated SM marker gene expression during postnatal vessel growth and after vascular injury. We also show that FRNK expression is regulated by transforming growth factor (TGF)-beta and that forced expression of FRNK in cultured cells induces serum- and TGF-beta-stimulated SM marker gene expression, whereas FRNK deletion or expression of a constitutively activated FAK variant attenuated SM gene transcription. CONCLUSIONS These data highlight the possibility that extrinsic signals regulate the SMC gene profile, at least in part, by modulating the expression of FRNK and that tight regulation of FAK activity by FRNK is important for proper SMC differentiation during development and after vascular injury.
Collapse
Affiliation(s)
- Rebecca L. Sayers
- Department of Physiology, University of North Carolina, Chapel Hill, NC 27599
| | | | - Mauricio Rojas
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599
| | - Haruko Hayasaka
- Department of Microbiology, University of Virginia Health System, Charlottesville, VA 22908
| | - J. Thomas Parsons
- Department of Microbiology, University of Virginia Health System, Charlottesville, VA 22908
| | - Christopher P. Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599
| | - Joan M. Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
45
|
Huang H, Asimaki A, Lo D, McKenna W, Saffitz J. Disparate effects of different mutations in plakoglobin on cell mechanical behavior. CELL MOTILITY AND THE CYTOSKELETON 2008; 65:964-78. [PMID: 18937352 PMCID: PMC2650236 DOI: 10.1002/cm.20319] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mutations in genes encoding desmosomal proteins have been implicated in the pathogenesis of heart and skin diseases. This has led to the hypothesis that defective cell-cell adhesion is the underlying cause of injury in tissues that repeatedly bear high mechanical loads. In this study, we examined the effects of two different mutations in plakoglobin on cell migration, stiffness, and adhesion. One is a C-terminal mutation causing Naxos disease, a recessive syndrome of arrhythmogenic right ventricular cardiomyopathy (ARVC) and abnormal skin and hair. The other is an N-terminal mutation causing dominant inheritance of ARVC without cutaneous abnormalities. To assess the effects of plakoglobin mutations on a broad range of cell mechanical behavior, we characterized a model system consisting of stably transfected HEK cells which are particularly well suited for analyses of cell migration and adhesion. Both mutations increased the speed of wound healing which appeared to be related to increased cell motility rather than increased cell proliferation. However, the C-terminal mutation led to dramatically decreased cell-cell adhesion, whereas the N-terminal mutation caused a decrease in cell stiffness. These results indicate that different mutations in plakoglobin have markedly disparate effects on cell mechanical behavior, suggesting complex biomechanical roles for this protein.
Collapse
Affiliation(s)
- Hayden Huang
- Department of Medicine, Brigham and Women's Hospital, Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Severe heart failure is associated with damage to the myocardium that is irreversible with current medical therapies. Recent experimental and clinical studies, however, have opened the possibility of solving many of the associated problems, making this an exciting and tangible goal. There are many potential cell sources for regenerative cardiac medicine, including bone marrow stem cells, endothelial progenitor cells, skeletal myocytes, adult cardiac stem cells, and embryonic stem (ES) cells. Although ES cells are highly proliferative and suitable for mass production, they are not autologous, and an efficient protocol is yet to be established to ensure selective cardiomyocyte induction. Recent studies have successfully established inducible pluripotent stem (iPS) cells from mouse and human fibroblasts by the gene transfer of 4 transcription factors that are strongly expressed in ES cells: Oct3/4, Sox2, Klf4 and c-Myc. iPS cells can differentiate into all 3 germ layer-derived cells and are syngeneic, indicating that they can become an ideal cell source for regenerative medicine. Despite these successes, the accumulating evidence from fields as diverse as developmental biology, stem cell biology and tissue engineering must be integrated to achieve the full potential of cardiac regenerative medicine.
Collapse
Affiliation(s)
- Shinsuke Yuasa
- Department of Regenerative Medicine and Advanced Cardiac Therapeutics, Keio University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
47
|
Abstract
Mechanical forces participate in morphogenesis from the level of individual cells to whole organism patterning. This article reviews recent research that has identified specific roles for mechanical forces in important developmental events. One well defined example is that dynein-driven cilia create fluid flow that determines left-right patterning in the early mammalian embryo. Fluid flow is also important for vasculogenesis, and evidence suggests that fluid shear stress rather than fluid transport is primarily required for remodeling the early vasculature. Contraction of the actin cytoskeleton, driven by nonmuscle myosins and regulated by the Rho family GTPases, is a recurring mechanism for controlling morphogenesis throughout development, from gastrulation to cardiogenesis. Finally, novel experimental approaches suggest critical roles for the actin cytoskeleton and the mechanical environment in determining differentiation of mesenchymal stem cells. Insights into the mechanisms linking mechanical forces to cell and tissue differentiation pathways are important for understanding many congenital diseases and for developing regenerative medicine strategies.
Collapse
Affiliation(s)
- Parth Patwari
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
48
|
Abstract
Pluripotent ES (embryonic stem) cells can be expanded in culture and induced to differentiate into a wide range of cell types. Self-renewal of ES cells involves proliferation with concomitant suppression of differentiation. Some critical and conserved pathways regulating self-renewal in both human and mouse ES cells have been identified, but there is also evidence suggesting significant species differences. Cytoplasmic and receptor tyrosine kinases play important roles in proliferation, survival, self-renewal and differentiation in stem, progenitor and adult cells. The present review focuses on the role of tyrosine kinase signalling for maintenance of the undifferentiated state, proliferation, survival and early differentiation of ES cells.
Collapse
Affiliation(s)
- Cecilia Annerén
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
49
|
Bauwens CL, Peerani R, Niebruegge S, Woodhouse KA, Kumacheva E, Husain M, Zandstra PW. Control of human embryonic stem cell colony and aggregate size heterogeneity influences differentiation trajectories. Stem Cells 2008; 26:2300-10. [PMID: 18583540 DOI: 10.1634/stemcells.2008-0183] [Citation(s) in RCA: 335] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To better understand endogenous parameters that influence pluripotent cell differentiation we used human embryonic stem cells (hESCs) as a model system. We demonstrate that differentiation trajectories in aggregate (embryoid body [EB])-induced differentiation, a common approach to mimic some of the spatial and temporal aspects of in vivo development, are affected by three factors: input hESC composition, input hESC colony size, and EB size. Using a microcontact printing approach, size-specified hESC colonies were formed by plating single-cell suspensions onto micropatterned (MP) extracellular matrix islands. Subsequently, size-controlled EBs were formed by transferring entire colonies into suspension culture enabling the independent investigation of colony and aggregate size effects on differentiation induction. Gene and protein expression analysis of MP-hESC populations revealed that the ratio of Gata6 (endoderm-associated marker) to Pax6 (neural-associated marker) expression increased with decreasing colony size. Moreover, upon forming EBs from these MP-hESCs, we observed that differentiation trajectories were affected by both colony and EB size-influenced parameters. In MP-EBs generated from endoderm-biased (high Gata6/Pax6) input hESCs, higher mesoderm and cardiac induction was observed at larger EB sizes. Conversely, neural-biased (low Gata6/Pax6) input hESCs generated MP-EBs that exhibited higher cardiac induction in smaller EBs. Our analysis demonstrates that heterogeneity in hESC colony and aggregate size, typical in most differentiation strategies, produces subsets of appropriate conditions for differentiation into specific cell types. Moreover, our findings suggest that the local microenvironment modulates endogenous parameters that can be used to influence pluripotent cell differentiation trajectories.
Collapse
Affiliation(s)
- Céline Liu Bauwens
- Department of Chemical Engineering and Applied Chemistry, University Health Network and Heart & Stroke Richard Lewar Centre of Excellence, University of Toronto,Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
50
|
Daley WP, Peters SB, Larsen M. Extracellular matrix dynamics in development and regenerative medicine. J Cell Sci 2008; 121:255-64. [PMID: 18216330 DOI: 10.1242/jcs.006064] [Citation(s) in RCA: 686] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix (ECM) regulates cell behavior by influencing cell proliferation, survival, shape, migration and differentiation. Far from being a static structure, the ECM is constantly undergoing remodeling--i.e. assembly and degradation--particularly during the normal processes of development, differentiation and wound repair. When misregulated, this can contribute to disease. ECM assembly is regulated by the 3D environment and the cellular tension that is transmitted through integrins. Degradation is controlled by complex proteolytic cascades, and misregulation of these results in ECM damage that is a common component of many diseases. Tissue engineering strives to replace damaged tissues with stem cells seeded on synthetic structures designed to mimic the ECM and thus restore the normal control of cell function. Stem cell self-renewal and differentiation is influenced by the 3D environment within the stem cell niche. For tissue-engineering strategies to be successful, the intimate dynamic relationship between cells and the ECM must be understood to ensure appropriate cell behavior.
Collapse
Affiliation(s)
- William P Daley
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | | | | |
Collapse
|