1
|
Xia JB, Liu K, Lin XL, Li HJ, Lin JH, Li L, Liang CQ, Cao Y, Wen N, Liao ZF, Zhao H, Park KS, Song GH, Ye ZB, Cai DQ, Ju ZY, Qi XF. FoxO3 controls cardiomyocyte proliferation and heart regeneration by regulating Sfrp2 expression in postnatal mice. Nat Commun 2025; 16:2532. [PMID: 40087279 PMCID: PMC11909131 DOI: 10.1038/s41467-025-57962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
The Forkhead box O3 (FoxO3) transcription factor is crucial to controlling heart growth in adulthood, but its exact role in cardiac repair and regeneration in postnatal mice remains unclear. Here, we show that FoxO3 deficiency promotes cardiomyocyte proliferation in postnatal mice and improves cardiac function in homeostatic adult mice. Moreover, FoxO3 deficiency accelerates heart regeneration following injury in postnatal mice at the regenerative and non-regenerative stages. We reveal that FoxO3 directly promotes the expression of secreted frizzled-related protein 2 (Sfrp2) and suppresses the activation of canonical Wnt/β-catenin signaling during heart regeneration. The increased activation of β-catenin in FoxO3-deficient cardiomyocytes can be blocked by Sfrp2 overexpression. In addition, Sfrp2 overexpression suppressed cardiomyocyte proliferation and heart regeneration in FoxO3-deficient mice. These findings suggest that FoxO3 negatively controls cardiomyocyte proliferation and heart regeneration in postnatal mice at least in part by promoting Sfrp2 expression, which leading to the inactivation of canonical Wnt/β-catenin signaling.
Collapse
Grants
- 82370247, 82070257, and 81770240 National Natural Science Foundation of China (National Science Foundation of China)
- the Fundamental Research Funds for the Central Universities (21623110), the Open Program of Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics (GPKLMMD-OP202302), the Research Grant of Key Laboratory of Regenerative Medicine of Ministry of Education (ZSYXM202402, ZSYXM202303, ZSYXM202206, and ZSYXM202104), the Guangdong Natural Science Funds for Distinguished Young Scholar (2014A030306011), and the Top Young Talents of Guangdong Province Special Support Program (87315007), China.
Collapse
Affiliation(s)
- Jing-Bo Xia
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510317, China
| | - Kun Liu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- Department of Cardiology, Zhongshan Torch Development Zone People's Hospital, Zhongshan, 528437, China
| | - Xiao-Lin Lin
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Hong-Ji Li
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jin-Hua Lin
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Chi-Qian Liang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yan Cao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Na Wen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhao-Fu Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523808, China
| | - Hui Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kyu-Sang Park
- Department of Physiology, Wonju College of Medicine, Yonsei University, Wonju, Gangwon, 220-701, Korea
| | - Guo-Hua Song
- School of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, 250117, China
| | - Ze-Bing Ye
- Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510317, China.
| | - Dong-Qing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Zhen-Yu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xu-Feng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
2
|
Zhang R, Wu M, Xiang D, Zhu J, Zhang Q, Zhong H, Peng Y, Wang Z, Ma G, Li G, Liu F, Ye W, Shi R, Zhou X, Babarinde IA, Su H, Chen J, Zhang X, Qin D, Hutchins AP, Pei D, Li D. A primate-specific endogenous retroviral envelope protein sequesters SFRP2 to regulate human cardiomyocyte development. Cell Stem Cell 2024; 31:1298-1314.e8. [PMID: 39146934 DOI: 10.1016/j.stem.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/04/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024]
Abstract
Endogenous retroviruses (ERVs) occupy a significant part of the human genome, with some encoding proteins that influence the immune system or regulate cell-cell fusion in early extra-embryonic development. However, whether ERV-derived proteins regulate somatic development is unknown. Here, we report a somatic developmental function for the primate-specific ERVH48-1 (SUPYN/Suppressyn). ERVH48-1 encodes a fragment of a viral envelope that is expressed during early embryonic development. Loss of ERVH48-1 led to impaired mesoderm and cardiomyocyte commitment and diverted cells to an ectoderm-like fate. Mechanistically, ERVH48-1 is localized to sub-cellular membrane compartments through a functional N-terminal signal peptide and binds to the WNT antagonist SFRP2 to promote its polyubiquitination and degradation, thus limiting SFRP2 secretion and blocking repression of WNT/β-catenin signaling. Knockdown of SFRP2 or expression of a chimeric SFRP2 with the ERVH48-1 signal peptide rescued cardiomyocyte differentiation. This study demonstrates how ERVH48-1 modulates WNT/β-catenin signaling and cell type commitment in somatic development.
Collapse
Affiliation(s)
- Ran Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Menghua Wu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Dan Xiang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Hong Kong Institute of Science & Innovation, Guangzhou Institutes of Biomedicine and Health, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jieying Zhu
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Hong Kong Institute of Science & Innovation, Guangzhou Institutes of Biomedicine and Health, Guangzhou, Guangdong 510530, China
| | - Qi Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Hui Zhong
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Hong Kong Institute of Science & Innovation, Guangzhou Institutes of Biomedicine and Health, Guangzhou, Guangdong 510530, China
| | - Yuling Peng
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Zhenhua Wang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Gang Ma
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guihuan Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Fengping Liu
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; Faculty of Medicine, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Weipeng Ye
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China
| | - Ruona Shi
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuemeng Zhou
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Isaac A Babarinde
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jiekai Chen
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Hong Kong Institute of Science & Innovation, Guangzhou Institutes of Biomedicine and Health, Guangzhou, Guangdong 510530, China
| | - Xiaofei Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Hong Kong Institute of Science & Innovation, Guangzhou Institutes of Biomedicine and Health, Guangzhou, Guangdong 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| | - Andrew P Hutchins
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou 310024, China.
| | - Dongwei Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China.
| |
Collapse
|
3
|
Cao M, Jiang X, Wang X, Gao P, Zou Y. Increased Secreted Frizzled-Related Protein 2 in Hypertension-Induced Left Ventricular Remodeling. Rev Cardiovasc Med 2024; 25:171. [PMID: 39076470 PMCID: PMC11267183 DOI: 10.31083/j.rcm2505171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 07/31/2024] Open
Abstract
Background Secreted frizzled-related protein 2 (sFRP2) is involved in various cardiovascular diseases. However, its relevance in left ventricular (LV) remodeling in patients with hypertension (HTN) is obscure. Methods In this study, 196 patients with HTN were included, 59 with echocardiographic LV remodeling. A total of 100 healthy subjects served as normal controls. The serum-sFRP2 level was measured by enzyme-linked immunosorbent assay (ELISA). Data were collected from medical records for baseline characteristics, biochemistry tests, and echocardiography. Receiver operating characteristic (ROC) curves were used to assess the distinguishing value of sFRP2 for LV remodeling in patients with HTN. Spearman rank correlation analysis was utilized to identify factors correlated with sFRP2. Cardiac sFRP2 was determined by Western blot and quantitative polymerase chain reaction (qPCR). Results The level of serum-sFRP2 was higher in HTN patients with echocardiographic LV remodeling than their non-remodeling counterparts. ROC analysis showed that the area under the curve (AUC) for sFRP2 in distinguishing echocardiographic LV remodeling in HTN patients was 0.791 (95% confidence interval (CI): 0.714-0.869). The sFRP2 was negatively correlated with LV dimension and positively correlated with relative wall thickness (RWT). The expression of sFRP2 was higher in hypertrophic hearts, which could be reversed by myricetin. Conclusions The serum level and cardiac sFRP2 increased in the setting of LV remodeling and decreased by myricetin. Serum sFRP2 may be a promising distinguishing factor for LV remodeling in HTN patients.
Collapse
Affiliation(s)
- Mengying Cao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Institute of Biomedical Sciences, Fudan University, 200032 Shanghai, China
| | - Xueli Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Institute of Biomedical Sciences, Fudan University, 200032 Shanghai, China
| | - Xiaolin Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Institute of Biomedical Sciences, Fudan University, 200032 Shanghai, China
| | - Pan Gao
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Institute of Biomedical Sciences, Fudan University, 200032 Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Institute of Biomedical Sciences, Fudan University, 200032 Shanghai, China
| |
Collapse
|
4
|
Ishioka M, Nihashi Y, Sunagawa Y, Umezawa K, Shimosato T, Kagami H, Morimoto T, Takaya T. Myogenetic Oligodeoxynucleotide Induces Myocardial Differentiation of Murine Pluripotent Stem Cells. Int J Mol Sci 2023; 24:14380. [PMID: 37762684 PMCID: PMC10532123 DOI: 10.3390/ijms241814380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
An 18-base myogenetic oligodeoxynucleotide (myoDN), iSN04, acts as an anti-nucleolin aptamer and induces myogenic differentiation of skeletal muscle myoblasts. This study investigated the effect of iSN04 on murine embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). In the undifferentiated state, iSN04 inhibited the proliferation of ESCs and iPSCs but did not affect the expression of pluripotent markers. In the differentiating condition, iSN04 treatment of ESCs/iPSCs from day 5 onward dramatically induced differentiation into Nkx2-5+ beating cardiomyocytes with upregulation of Gata4, Isl1, and Nkx2-5, whereas iSN04 treatment from earlier stages completely inhibited cardiomyogenesis. RNA sequencing revealed that iSN04 treatment from day 5 onward contributes to the generation of cardiac progenitors by modulating the Wnt signaling pathway. Immunostaining showed that iSN04 suppressed the cytoplasmic translocation of nucleolin and restricted it to the nucleoli. These results demonstrate that nucleolin inhibition by iSN04 facilitates the terminal differentiation of cardiac mesoderm into cardiomyocytes but interferes with the differentiation of early mesoderm into the cardiac lineage. This is the first report on the generation of cardiomyocytes from pluripotent stem cells using a DNA aptamer. Since iSN04 did not induce hypertrophic responses in primary-cultured cardiomyocytes, iSN04 would be useful and safe for the regenerative therapy of heart failure using stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Mina Ishioka
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
| | - Yuma Nihashi
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 5-41, 1-1-1 Higashi, Tsukuba 305-8565, Ibaraki, Japan;
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.S.); (T.M.)
| | - Koji Umezawa
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Takeshi Shimosato
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| | - Hiroshi Kagami
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (Y.S.); (T.M.)
| | - Tomohide Takaya
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (M.I.); (T.S.)
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan; (K.U.); (H.K.)
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-minowa, Kami-ina, Nagano 399-4598, Japan
| |
Collapse
|
5
|
Bryl R, Nawrocki MJ, Jopek K, Kaczmarek M, Bukowska D, Antosik P, Mozdziak P, Zabel M, Dzięgiel P, Kempisty B. Transcriptomic Characterization of Genes Regulating the Stemness in Porcine Atrial Cardiomyocytes during Primary In Vitro Culture. Genes (Basel) 2023; 14:1223. [PMID: 37372403 PMCID: PMC10297922 DOI: 10.3390/genes14061223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Heart failure remains a major cause of death worldwide. There is a need to establish new management options as current treatment is frequently suboptimal. Clinical approaches based on autologous stem cell transplant is potentially a good alternative. The heart was long considered an organ unable to regenerate and renew. However, several reports imply that it may possess modest intrinsic regenerative potential. To allow for detailed characterization of cell cultures, whole transcriptome profiling was performed after 0, 7, 15, and 30 days of in vitro cell cultures (IVC) from the right atrial appendage and right atrial wall utilizing microarray technology. In total, 4239 differentially expressed genes (DEGs) with ratio > abs |2| and adjusted p-value ≤ 0.05 for the right atrial wall and 4662 DEGs for the right atrial appendage were identified. It was shown that a subset of DEGs, which have demonstrated some regulation of expression levels with the duration of the cell culture, were enriched in the following GO BP (Gene Ontology Biological Process) terms: "stem cell population maintenance" and "stem cell proliferation". The results were validated by RT-qPCR. The establishment and detailed characterization of in vitro culture of myocardial cells may be important for future applications of these cells in heart regeneration processes.
Collapse
Affiliation(s)
- Rut Bryl
- Section of Regenerative Medicine and Cancer Research, Natural Sciences Club, Faculty of Biology, Adam Mickiewicz University, Poznań, 61-614 Poznan, Poland;
| | - Mariusz J. Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland;
- Gene Therapy Laboratory, Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
| | - Maciej Zabel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Piotr Dzięgiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.Z.); (P.D.)
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Department of Obstetrics and Gynaecology, University Hospital and Masaryk University, 62500 Brno, Czech Republic
| |
Collapse
|
6
|
Yamamoto T, Kambayashi Y, Otsuka Y, Afouda B, Giuraniuc C, Michiue T, Hoppler S. Positive feedback regulation of frizzled-7 expression robustly shapes a steep Wnt gradient in Xenopus heart development, together with sFRP1 and heparan sulfate. eLife 2022; 11:73818. [PMID: 35942683 PMCID: PMC9363125 DOI: 10.7554/elife.73818] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Secreted molecules called morphogens govern tissue patterning in a concentration-dependent manner. However, it is still unclear how reproducible patterning can be achieved with diffusing molecules, especially when that patterning concerns differentiation of thin tissues. Wnt is a morphogen that organizes cardiac development. Wnt6 patterns cardiogenic mesoderm to induce differentiation of a thin tissue, the pericardium, in Xenopus. In this study, we revealed that a Wnt receptor, frizzled-7, is expressed in a Wnt-dependent manner. With a combination of experiments and mathematical modeling, this receptor-feedback appears essential to shape a steep gradient of Wnt signaling. In addition, computer simulation revealed that this feedback imparts robustness against variations of Wnt ligand production and allows the system to reach a steady state quickly. We also found that a Wnt antagonist sFRP1, which is expressed on the opposite side of the Wnt source, accumulates on N-acetyl-rich heparan sulfate (HS). N-acetyl-rich HS concentration is high between the sources of Wnt and sFRP1, achieving local inhibition of Wnt signaling via restriction of sFRP1 spreading. These integrated regulatory systems restrict the Wnt signaling range and ensure reproducible patterning of the thin pericardium.
Collapse
Affiliation(s)
- Takayoshi Yamamoto
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo
| | - Yuta Kambayashi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo
| | - Yuta Otsuka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo
| | - Boni Afouda
- Institute of Medical Sciences, The University of Aberdeen
| | | | - Tatsuo Michiue
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo
| | - Stefan Hoppler
- Institute of Medical Sciences, The University of Aberdeen
| |
Collapse
|
7
|
The glycoprotein Wnt6 regulates human dental papilla cells differentiation by canonical Wnt signaling pathway. Arch Oral Biol 2022; 141:105469. [DOI: 10.1016/j.archoralbio.2022.105469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
|
8
|
Bonnet C, González S, Roberts JS, Robertson SYT, Ruiz M, Zheng J, Deng SX. Human limbal epithelial stem cell regulation, bioengineering and function. Prog Retin Eye Res 2021; 85:100956. [PMID: 33676006 PMCID: PMC8428188 DOI: 10.1016/j.preteyeres.2021.100956] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
The corneal epithelium is continuously renewed by limbal stem/progenitor cells (LSCs), a cell population harbored in a highly regulated niche located at the limbus. Dysfunction and/or loss of LSCs and their niche cause limbal stem cell deficiency (LSCD), a disease that is marked by invasion of conjunctival epithelium into the cornea and results in failure of epithelial wound healing. Corneal opacity, pain, loss of vision, and blindness are the consequences of LSCD. Successful treatment of LSCD depends on accurate diagnosis and staging of the disease and requires restoration of functional LSCs and their niche. This review highlights the major advances in the identification of potential LSC biomarkers and components of the LSC niche, understanding of LSC regulation, methods and regulatory standards in bioengineering of LSCs, and diagnosis and staging of LSCD. Overall, this review presents key points for researchers and clinicians alike to consider in deepening the understanding of LSC biology and improving LSCD therapies.
Collapse
Affiliation(s)
- Clémence Bonnet
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA; Cornea Department, Paris University, Cochin Hospital, AP-HP, F-75014, Paris, France
| | - Sheyla González
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - JoAnn S Roberts
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Sarah Y T Robertson
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Maxime Ruiz
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Jie Zheng
- Basic Science Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA
| | - Sophie X Deng
- Cornea Division, Stein Eye Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
9
|
Cao M, Wang H, Li W, Jiang X, Wang X, Guo W, Gao P, Zou Y. Inverse Associations Between Circulating Secreted Frizzled Related Protein 2 (sFRP2) and Cardiometabolic Risk Factors. Front Cardiovasc Med 2021; 8:723205. [PMID: 34722660 PMCID: PMC8551478 DOI: 10.3389/fcvm.2021.723205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/22/2021] [Indexed: 12/05/2022] Open
Abstract
Background: Secreted frizzled-related protein 2 (sFRP2) plays an important role in metabolic syndrome and cardiovascular diseases (CVDs); However, its relevance with cardiometabolic diseases remains to be elucidated. We aimed to determine the serum levels of sFRP2 in patients at different stages of heart failure (HF) with or without type 2 diabetes mellitus (T2DM), and assess the correlation between circulating sFRP2 levels and cardiometabolic risk factors. Methods: In this study, serum samples from 277 patients visiting Zhongshan Hospital affiliated to Fudan University were collected. These patients were clinically diagnosed and categorized as five groups, including the control group, pre-clinical HF group, pre-clinical HF+T2DM group, HF group and HF+T2DM group. Serum sFRP2 levels were measured with enzyme-linked immunosorbent assay (ELISA) tests and the clinical characteristics of each patient were recorded. Spearman rank correlation analysis and multiple stepwise linear regression analysis were conducted. Univariate and multivariate logistic regression analysis were performed to screen risk factors for HF in patients with CVDs. Results: Serum sFRP2 levels were significantly lower in the HF+T2DM group compared with the other four groups. Spearman rank correlation analysis showed that sFRP2 was negatively correlated with parameters including patients' age, fasting plasma glucose (FPG), glycated hemoglobin A1c (HbA1c), cardiac troponin T (cTNT), N-terminal pro-B-type natriuretic peptide (NT-proBNP), high-sensitivity C-reactive protein (hs-CRP), left atrial dimension (LAD) and left ventricular posterior wall (LVPW), and positively correlated with hemoglobin, estimated glomerular filtration rate (eGFR), albumin, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and left ventricular ejection fraction (LVEF). However, in multiple regression analysis, significant associations with ln(sFRP2) were observed only in FPG, hs-CRP and LAD. Higher serum sFRP2 was significantly linked to lower odds of HF in patients with CVDs. Conclusion: sFRP2 progressively decreased when glucose homeostasis and cardiac function deteriorated. sFRP2 acted as a risk factor for HF in patients with CVDs, especially in those with concomitant T2DM.
Collapse
Affiliation(s)
- Mengying Cao
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenshu Li
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and Key Lab of Health Technology Assessment of the Ministry of Health, Fudan University, Shanghai, China
| | - Xueli Jiang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaolin Wang
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Pan Gao
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Shanghai Clinical Bioinformatics Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Mesenchymal Stem Cell Transplantation for the Treatment of Age-Related Musculoskeletal Frailty. Int J Mol Sci 2021; 22:ijms221910542. [PMID: 34638883 PMCID: PMC8508885 DOI: 10.3390/ijms221910542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 12/16/2022] Open
Abstract
Projected life expectancy continues to grow worldwide owing to the advancement of new treatments and technologies leading to rapid growth of geriatric population. Thus, age-associated diseases especially in the musculoskeletal system are becoming more common. Loss of bone (osteoporosis) and muscle (sarcopenia) mass are conditions whose prevalence is increasing because of the change in population distribution in the world towards an older mean age. The deterioration in the bone and muscle functions can cause severe disability and seriously affects the patients’ quality of life. Currently, there is no treatment to prevent and reverse age-related musculoskeletal frailty. Existing interventions are mainly to slow down and control the signs and symptoms. Mesenchymal stem cell (MSC) transplantation is a promising approach to attenuate age-related musculoskeletal frailty. This review compiles the present knowledge of the causes and changes of the musculoskeletal frailty and the potential of MSC transplantation as a regenerative therapy for age-related musculoskeletal frailty.
Collapse
|
11
|
Merimi M, El-Majzoub R, Lagneaux L, Moussa Agha D, Bouhtit F, Meuleman N, Fahmi H, Lewalle P, Fayyad-Kazan M, Najar M. The Therapeutic Potential of Mesenchymal Stromal Cells for Regenerative Medicine: Current Knowledge and Future Understandings. Front Cell Dev Biol 2021; 9:661532. [PMID: 34490235 PMCID: PMC8416483 DOI: 10.3389/fcell.2021.661532] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022] Open
Abstract
In recent decades, research on the therapeutic potential of progenitor cells has advanced considerably. Among progenitor cells, mesenchymal stromal cells (MSCs) have attracted significant interest and have proven to be a promising tool for regenerative medicine. MSCs are isolated from various anatomical sites, including bone marrow, adipose tissue, and umbilical cord. Advances in separation, culture, and expansion techniques for MSCs have enabled their large-scale therapeutic application. This progress accompanied by the rapid improvement of transplantation practices has enhanced the utilization of MSCs in regenerative medicine. During tissue healing, MSCs may exhibit several therapeutic functions to support the repair and regeneration of injured tissue. The process underlying these effects likely involves the migration and homing of MSCs, as well as their immunotropic functions. The direct differentiation of MSCs as a cell replacement therapeutic mechanism is discussed. The fate and behavior of MSCs are further regulated by their microenvironment, which may consequently influence their repair potential. A paracrine pathway based on the release of different messengers, including regulatory factors, chemokines, cytokines, growth factors, and nucleic acids that can be secreted or packaged into extracellular vesicles, is also implicated in the therapeutic properties of MSCs. In this review, we will discuss relevant outcomes regarding the properties and roles of MSCs during tissue repair and regeneration. We will critically examine the influence of the local microenvironment, especially immunological and inflammatory signals, as well as the mechanisms underlying these therapeutic effects. Importantly, we will describe the interactions of local progenitor and immune cells with MSCs and their modulation during tissue injury. We will also highlight the crucial role of paracrine pathways, including the role of extracellular vesicles, in this healing process. Moreover, we will discuss the therapeutic potential of MSCs and MSC-derived extracellular vesicles in the treatment of COVID-19 (coronavirus disease 2019) patients. Overall, this review will provide a better understanding of MSC-based therapies as a novel immunoregenerative strategy.
Collapse
Affiliation(s)
- Makram Merimi
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
- LBBES Laboratory, Genetics and Immune-Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Rania El-Majzoub
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Beirut, Lebanon
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Douâa Moussa Agha
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Fatima Bouhtit
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
- LBBES Laboratory, Genetics and Immune-Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Nathalie Meuleman
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Philippe Lewalle
- Laboratory of Experimental Hematology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Bruxelles, Belgium
| | - Mohammad Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
12
|
Abstract
Cardiomyogenesis, the process by which the body generates cardiomyocytes, is poorly understood. We have recently shown that Sfrp2 promotes cardiomyogenesis in vitro. The objective of this study was to determine if Sfrp2 would similarly promote cardiomyogenesis in vivo. To test this hypothesis, we tracked multipotent cKit(+) cells in response to Sfrp2 treatment. In control adult mice, multipotent cKit(+) cells typically differentiated into endothelial cells but not cardiomyocytes. In contrast, Sfrp2 switched the fate of these cells. Following Sfrp2 injection, multipotent cKit(+) cells differentiated solely into cardiomyocytes. Sfrp2-derived cardiomyocytes integrated into the myocardium and exhibited identical physiological properties to preexisting native cardiomyocytes. The ability of Sfrp2 to promote cardiomyogenesis was further supported by tracking EdU-labeled cells. In addition, Sfrp2 did not promote the formation of new cardiomyocytes when the cKit(+) cell population was selectively ablated in vivo using a diphtheria toxin receptor-diphtheria toxin model. Notably, Sfrp2-induced cardiomyogenesis was associated with significant functional improvements in a cardiac injury model. In summary, our study further demonstrates the importance of Sfrp2 in cardiomyogenesis.
Collapse
|
13
|
Zurbrigg K, Bertolini F, Walugembe M, van Dreumel T, Alves D, Friendship R, O'Sullivan TL, Rothschild MF. A genome-wide analysis of cardiac lesions of pigs that die during transport: Is heart failure of in-transit-loss pigs associated with a heritable cardiomyopathy? CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2021; 85:119-126. [PMID: 33883819 PMCID: PMC7995549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/25/2020] [Indexed: 06/12/2023]
Abstract
While heart failure is a primary cause of death for many in-transit-loss (ITL) pigs, the underlying cause of these deaths is not known. Cardiomyopathies are considered a common cause of heart failure in humans and often have a genetic component. The objective of this study was to determine if genes associated with cardiomyopathies could be identified in ITL pigs. Samples from the hearts of pigs that died during transport to an abattoir in Ontario, Canada were collected and genotyped along with samples from pigs that did not die during transport (ILT hearts: n = 149; non-ITL/control hearts: n = 387). Genome-wide analyses were carried out on each of the determined phenotypes (gross cardiac lesions) using a medium density single nucleotide polymorphism (SNP) chip and 500 kb windows/regions for analysis, with 250 kb regions of overlap. The distribution derived by a multidimensional scaling (MDS) analysis of all phenotypes demonstrated a lack of complete separation between phenotypes of affected and unaffected animals, which made diagnosis difficult. Although genetic differences were small, a few genes associated with dilated cardiomyopathy (DCM) and arrhythmogenic right ventricular cardiomyopathy (ARVM) were identified. In addition, multiple genes associated with cardiac arrhythmias and ventricular hypertrophy were identified that can possibly result in heart failure. The results of this preliminary study did not provide convincing evidence that a single, heritable cardiomyopathy is the cause of heart failure in ITL pigs.
Collapse
Affiliation(s)
- Katherine Zurbrigg
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Francesca Bertolini
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Muhammed Walugembe
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Toni van Dreumel
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - David Alves
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Robert Friendship
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Terri L O'Sullivan
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| | - Max F Rothschild
- Department of Population Medicine, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1 (Zurbrigg, Friendship, O'Sullivan); Department of Animal Science, Iowa State University, Ames, Iowa 50011, USA (Bertolini, Walugembe, Rothschild); National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, Denmark (Bertolini); Veterinary Pathology Consultant, Ontario (van Dreumel), Veterinary Epidemiology Consultant, Elora, Ontario (Alves)
| |
Collapse
|
14
|
Hsueh YC, Hodgkinson CP, Gomez JA. The role of Sfrp and DKK proteins in cardiomyocyte development. Physiol Rep 2021; 9:e14678. [PMID: 33587322 PMCID: PMC7883806 DOI: 10.14814/phy2.14678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, we summarize the role of Wnt proteins in cardiomyogenesis. More specifically, we focus on how the development of cardiomyocytes from precursor cells involves a complex interplay between Wnt canonical β-catenin signaling pathways and Wnt noncanonical signaling pathways involving PCP and JNK. We also describe recent literature which suggests that endogenous Wnt inhibitors such as the Sfrp and DKK proteins play important roles in regulating the cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Ying-Chang Hsueh
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Jose A Gomez
- Department of Medicine, Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
15
|
He J, Jing J, Feng J, Han X, Yuan Y, Guo T, Pei F, Ma Y, Cho C, Ho TV, Chai Y. Lhx6 regulates canonical Wnt signaling to control the fate of mesenchymal progenitor cells during mouse molar root patterning. PLoS Genet 2021; 17:e1009320. [PMID: 33596195 PMCID: PMC7920342 DOI: 10.1371/journal.pgen.1009320] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/01/2021] [Accepted: 12/21/2020] [Indexed: 02/05/2023] Open
Abstract
Mammalian tooth crown formation has long served as a model for investigating how patterning and morphogenesis are orchestrated during development. However, the mechanism underlying root patterning and morphogenesis remains poorly understood. In this study, we find that Lhx6 labels a subpopulation of root progenitor cells in the apical dental mesenchyme, which is closely associated with furcation development. Loss of Lhx6 leads to furcation and root number defects, indicating that Lhx6 is a key root patterning regulator. Among the multiple cellular events regulated by Lhx6 is the odontoblast fate commitment of progenitor cells, which it controls in a cell-autonomous manner. Specifically, Lhx6 loss leads to elevated expression of the Wnt antagonist Sfrp2 and down-regulation of Wnt signaling in the furcation region, while overactivation of Wnt signaling in Lhx6+ progenitor cells partially restore the furcation defects in Lhx6-/- mice. Collectively, our findings have important implications for understanding organ morphogenesis and future strategies for tooth root regeneration.
Collapse
Affiliation(s)
- Jinzhi He
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan province, China
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Xia Han
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Yuan Yuan
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Tingwei Guo
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Fei Pei
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Yuanyuan Ma
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Courtney Cho
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
16
|
Wei M, Zhang C, Tian Y, Du X, Wang Q, Zhao H. Expression and Function of WNT6: From Development to Disease. Front Cell Dev Biol 2021; 8:558155. [PMID: 33425886 PMCID: PMC7794017 DOI: 10.3389/fcell.2020.558155] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/04/2020] [Indexed: 11/17/2022] Open
Abstract
WNT family member 6 (WNT6) is a member of the highly conserved WNT protein family. It plays an essential role in the normal development process, not only in embryonic morphogenesis, but also in post-natal homeostasis. WNT6 functions in mice and humans. This review summarizes the current findings on the biological functions of WNT6, describing its involvement in regulating embryogenesis, decidualization, and organ development. Aberrant WNT6 signaling is related to various pathologies, such as promoting cancer development, lung tuberculosis, and kidney fibrosis and improving the symptoms of Rett syndrome (RTT). Thus, due to its various functions, WNT6 has great potential for in-depth research. This work not only describes the signaling mechanism and function of WNT6 under physiological and pathological conditions, but also provides a theoretical basis for targeted therapy.
Collapse
Affiliation(s)
- Ming Wei
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Congmin Zhang
- Department of Scientific Research Center, The Second Hospital of Dalian Medical University, Dalian, China
| | - Yujia Tian
- Department of Scientific Research Center, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xiaohui Du
- Department of Scientific Research Center, The Second Hospital of Dalian Medical University, Dalian, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Hui Zhao
- The Health Check Up Center, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Huang A, Huang Y. Role of Sfrps in cardiovascular disease. Ther Adv Chronic Dis 2020; 11:2040622320901990. [PMID: 32064070 PMCID: PMC6987486 DOI: 10.1177/2040622320901990] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022] Open
Abstract
Secreted frizzled-related proteins (Sfrps) are a family of secreted proteins that bind extracellularly to Wnt ligands and frizzled receptors. This binding modulates the Wnt signaling cascade, and Sfrps interact with their corresponding receptors. Sfrps are thought to play an important role in the pathological mechanism of cardiac disease such as myocardial infarction, cardiac remodeling, and heart failure. However, the overall role of Sfrps in cardiac disease is unknown. Some members of the Sfrps family modulate cellular apoptosis, angiogenesis, differentiation, the inflammatory process, and cardiac remodeling. In this review, we summarize the evidence of Sfrps association with cardiac disease. We also discuss how multiple mechanisms may underlie Sfrps being involved in such diverse pathologies.
Collapse
Affiliation(s)
- Anqing Huang
- Department of Cardiology, Shunde Hospital,
Southern Medical University, Foshan, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital,
Southern Medical University, Jiazhi Road, Lunjiao Town, Shunde District,
Foshan, Guangdong 528300, China The George Institute for Global Health, NSW
2042, Australia
| |
Collapse
|
18
|
Wu Y, Liu X, Zheng H, Zhu H, Mai W, Huang X, Huang Y. Multiple Roles of sFRP2 in Cardiac Development and Cardiovascular Disease. Int J Biol Sci 2020; 16:730-738. [PMID: 32071544 PMCID: PMC7019133 DOI: 10.7150/ijbs.40923] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022] Open
Abstract
The Wnt signaling pathway plays important roles in organ development and disease processes. Secreted frizzled-related protein 2 (sFRP2), a vital molecule of Wnt signaling, can regulate cardiac development and cardiovascular disease. Recent studies have suggested that sFRP2 is not only an antagonist of the canonical Wnt signaling pathway, but also has a more complex relationship in myocardial fibrosis, angiogenesis, cardiac hypertrophy and cardiac regeneration. Here, we review the role of sFRP2 and Wnt signaling in cardiac development and cardiovascular disease.
Collapse
Affiliation(s)
- Yu Wu
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
| | - Xinyue Liu
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
| | - Hailan Zhu
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
| | - Weiyi Mai
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou
| | - Xiaohui Huang
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
| | - Yuli Huang
- Department of Cardiology, Shunde hospital, Southern Medical University, Jiazi Road 1 Lunjiao Town, Shunde District, Foshan, Guangdong, 528308, China
- The George Institute for Global Health, NSW 2042 Australia
| |
Collapse
|
19
|
González S, Oh D, Baclagon ER, Zheng JJ, Deng SX. Wnt Signaling Is Required for the Maintenance of Human Limbal Stem/Progenitor Cells In Vitro. Invest Ophthalmol Vis Sci 2019; 60:107-112. [PMID: 30640975 PMCID: PMC6333110 DOI: 10.1167/iovs.18-25740] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose A chemical approach to examine the role of Wnt signaling in maintaining the stemness and/or proliferation of limbal stem/progenitor cells (LSCs). Methods LSCs were isolated from human donor eyes and cultured as single cells for 12 to 14 days with the following small molecules: IIIC3, an antagonist of the Wnt signaling inhibitor Dickkopf (DKK), and IC15, a Wnt signaling inhibitor. Proliferation of LSCs in the presence of IIIC3 and IC15 was determined by the number of cells and colonies established. Maintenance of stemness was determined by p63α, cytokeratin (K)12, and K14 expression. Results Activation of Wnt, through IIIC3-mediated DKK inhibition, resulted in similar colony forming efficiency (CFE) as in the untreated LSCs, but significantly increased the number of cultivated cells 7.21% with 5 μM. Inhibition of Wnt with IC15 significantly reduced the CFE (P ≤ 0.01) and the number of cultivated cells by 16% to 29%. Percentage of cells expressing high levels of p63α (p63αbright) and quantity of small cells (≤12 μm), which contain the LSCs, increased 4.71% and 11.26% (both P < 0.05), respectively, with 5 μM IIIC3. All concentrations of IIIC3 and IC15 retained the K14 undifferentiated marker (97%), while differentiation, as detected by expression of K12, was found in up to 2% of cells in 1 μM IIIC3, 1 μM IC15, or 5 μM IIIC3. Conclusions Wnt signaling is required in LSC proliferation and maintenance of an undifferentiated state. The current study is a proof of concept that the Wnt pathway could be modulated in LSCs to enhance or decrease the efficiency of human LSC expansion.
Collapse
Affiliation(s)
- Sheyla González
- Stein Eye Institute, University of California, Los Angeles, California, United States
| | - Denise Oh
- Stein Eye Institute, University of California, Los Angeles, California, United States
| | - Elfren R Baclagon
- Stein Eye Institute, University of California, Los Angeles, California, United States
| | - Jie J Zheng
- Stein Eye Institute, University of California, Los Angeles, California, United States
| | - Sophie X Deng
- Stein Eye Institute, University of California, Los Angeles, California, United States
| |
Collapse
|
20
|
Kaplan NA, Wang W, Christiaen L. Initial characterization of Wnt-Tcf functions during Ciona heart development. Dev Biol 2019; 448:199-209. [PMID: 30635127 PMCID: PMC6487219 DOI: 10.1016/j.ydbio.2018.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/14/2018] [Accepted: 12/17/2018] [Indexed: 12/16/2022]
Abstract
In vertebrate embryos, the cardiopharyngeal mesoderm gives rise to both cardiac and branchiomeric head muscles. The canonical Wnt signaling pathway regulates many aspects of cardiomyocyte specification, and modulates a balance between skeletal and cardiac myogenesis during vertebrate head muscle development. However, the role of Wnt signaling during ascidian cardiopharyngeal development remains elusive. Here, we documented the expression of Wnt pathway components during cardiopharyngeal development in Ciona, and generated tools to investigate potential roles for Wnt signaling, and its transcriptional effector Tcf, on heart vs. pharyngeal muscle fate specification. Neither focused functional analyses nor lineage-specific transcriptome profiling uncovered a significant role for Tcf during early cardiac vs. pharyngeal muscle fate choice. By contrast, Wnt gene expression patterns of Frizzled4 and Lrp4/8 and CRISPR/Cas9-mediated Tcf knock-down suggested a later requirement for Wnt signaling during heart morphogenesis and/or cardiomyocyte differentiation. This study provides a provisional set of reagents to study Wnt signaling function in Ciona, and promising insights for future analyses of Wnt functions during heart organogenesis.
Collapse
Affiliation(s)
- Nicole A Kaplan
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Wei Wang
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA
| | - Lionel Christiaen
- Center for Developmental Genetics, Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
21
|
Sun XL, Hao QK, Tang RJ, Xiao C, Ge ML, Dong BR. Frailty and Rejuvenation with Stem Cells: Therapeutic Opportunities and Clinical Challenges. Rejuvenation Res 2019; 22:484-497. [PMID: 30693831 PMCID: PMC6919243 DOI: 10.1089/rej.2017.2048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Frailty, one appealing target for improving successful aging of the elderly population, is a common clinical syndrome based on the accumulation of multisystemic function declines and the increase in susceptibility to stressors during biological aging. The age-dependent senescence, the frailty-related stem cell depletion, chronic inflammation, imbalance of immune homeostasis, and the reduction of multipotent stem cells collectively suggest the rational hypothesis that it is possible to (partially) cure frailty with stem cells. This systematic review has included all of the human trials of stem cell therapy for frailty from the main electronic databases and printed materials and screened the closely related reviews themed on the mechanisms of aging, frailty, and stem cells, to provide more insights in stem cell strategies for frailty, one promising method to recover health from a frail status. To date, a total of four trials about this subject have been registered on clinicaltrials.gov. The use of mesenchymal stem cells (MSCs), doses of 100 million cells, single peripheral intravenous infusion, follow-up periods of 6–12 months, and a focus primarily on safety and secondarily on efficacy are common characteristics of these studies. We conclude that intravenous infusion of allogenic MSCs is safe, well tolerated, and preliminarily effective clinically. More preclinical experiments and clinical trials are warranted to precisely elucidate the mechanism, safety, and efficacy of frailty stem cell therapy.
Collapse
Affiliation(s)
- Xue-Lian Sun
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| | - Qiu-Kui Hao
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ren-Jie Tang
- Department of Urinary Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Urology, Chengdu Sixth People's Hospital, Chengdu, China
| | - Chun Xiao
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Sichuan Engineering Research Institute of Chi Ding Sheng Tong, Chengdu, China
| | - Mei-Ling Ge
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| | - Bi-Rong Dong
- National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, China
| |
Collapse
|
22
|
Henderson DJ, Long DA, Dean CH. Planar cell polarity in organ formation. Curr Opin Cell Biol 2018; 55:96-103. [PMID: 30015152 DOI: 10.1016/j.ceb.2018.06.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/07/2018] [Accepted: 06/18/2018] [Indexed: 01/11/2023]
Abstract
The planar cell polarity (PCP) pathway controls a variety of morphological events across many species. During embryonic development, the PCP pathway regulates coordinated behaviour of groups of cells to direct morphogenetic processes such as convergent extension and collective cell migration. In this review we discuss the increasingly prominent role of the PCP pathway in organogenesis, focusing on the lungs, kidneys and heart. We also highlight emerging evidence that PCP gene mutations are associated with adult diseases.
Collapse
Affiliation(s)
- Deborah J Henderson
- Cardiovascular Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Charlotte H Dean
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London, UK.
| |
Collapse
|
23
|
Insights from molecular signature of in vivo cardiac c-Kit(+) cells following cardiac injury and β-catenin inhibition. J Mol Cell Cardiol 2018; 123:64-74. [PMID: 30171847 DOI: 10.1016/j.yjmcc.2018.08.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/03/2018] [Accepted: 08/28/2018] [Indexed: 01/26/2023]
Abstract
There is much interest over resident c-Kit(+) cells in tissue regeneration. Their role in cardiac regeneration has been controversial. In this study we aim to understand the in vivo behavior of cardiac c-Kit(+) cells at baseline and after myocardial infarction and in response to Sfrp2. This approach can accurately study the in vivo transcript expressions of these cells in temporal response to injury and overcomes the limitations of the in vitro approach. RNA-seq was performed with c-Kit(+) cells and cardiomyocytes from healthy non-injured mice as well as c-Kit(+) cells from 1 day post-MI and 12 days post-MI mice. When compared to in vivo c-Kit(+) cells isolated from a healthy non-injured mouse heart, cardiomyocytes were enriched in transcripts that express anion channels, cation channels, developmental/differentiation pathway components, as well as proteins that inhibit canonical Wnt/β-catenin signaling. Myocardial infarction (MI) induced in vivo c-Kit(+) cells to transiently adopt the cardiomyocyte-specific signature: expression of a number of cardiomyocyte-specific transcripts was maximal 1 day post-MI and declined by 12 days post-MI. We next studied the effect of β-catenin inhibition on in vivo c-Kit(+) cells by administering the Wnt inhibitor Sfrp2 into the infarct border zone. Sfrp2 both enhanced and sustained cardiomyocyte-specific gene expression in the in vivo c-Kit(+) cells: expression of cardiomyocyte-specific transcripts was higher and there was no decline in expression by 12 days post-MI. Further analysis of the biology of c-Kit(+) cells identified that culture induced a significant and irreversible change in their molecular signature raising questions about reliability of in vitro studies. Our findings provide evidence that MI induces in vivo c-Kit(+) cells to adopt transiently a cardiomyocyte-specific pattern of gene expression, and Sfrp2 further enhances and induces sustained gene expression. Our approach is important for understanding c-Kit(+) cells in cardiac regeneration and also has broad implications in the investigation of in vivo resident stem cells in other areas of tissue regeneration.
Collapse
|
24
|
Teiken K, Kuehnel M, Rehkaemper J, Kreipe H, Laenger F, Hussein K, Jonigk D. Non-canonical WNT6/WNT10A signal factor expression in EBV+ post-transplant smooth muscle tumors. Clin Sarcoma Res 2018; 8:10. [PMID: 29881541 PMCID: PMC5985559 DOI: 10.1186/s13569-018-0096-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/07/2018] [Indexed: 12/11/2022] Open
Abstract
Post-transplant smooth muscle tumors (PTSMTs) are rare mesenchymal neoplasms which occur after solid organ or haematopoietic stem cell transplantation. PTSMT typically consist of Epstein–Barr-virus (EBV)+ smooth muscle-like cells and show an intermediate malignancy. Their main occurrences are visceral organs, especially the liver, but intracranial appearances are described and associated with a poor prognosis. EBV drives the growth of PTSMT; however, the underlying molecular mechanisms still remain unclear. Gene expression analysis of a set of morphologically similar tumors (leiomyomas, leiomyosarcomas, angioleiomyomas and endothelial haemangiomas) from patients without immunosuppression or EBV-association was performed. Our findings indicate that PTSMT’s growth is driven by two factors of the wingless-type protein family: WNT6 and WNT10A. We are first to report that in PTSMTs, a non-canonical activation of WNT, independent of beta-catenin, drives tumor cell proliferation via MTOR/AKT1, MYC and Cyclin D2.
Collapse
Affiliation(s)
- Kristin Teiken
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Mark Kuehnel
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Jan Rehkaemper
- 2Institute of Pathology, University of Muenster, Domagkstraße 17, 48149 Muenster, Germany
| | - Hans Kreipe
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Florian Laenger
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kais Hussein
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Danny Jonigk
- 1Institute of Pathology, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
25
|
Giancotti V, Bergamin N, Cataldi P, Rizzi C. Epigenetic Contribution of High-Mobility Group A Proteins to Stem Cell Properties. Int J Cell Biol 2018; 2018:3698078. [PMID: 29853899 PMCID: PMC5941823 DOI: 10.1155/2018/3698078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023] Open
Abstract
High-mobility group A (HMGA) proteins have been examined to understand their participation as structural epigenetic chromatin factors that confer stem-like properties to embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and cancer stem cells (CSCs). The function of HMGA was evaluated in conjunction with that of other epigenetic factors such as histones and microRNAs (miRs), taking into consideration the posttranscriptional modifications (PTMs) of histones (acetylation and methylation) and DNA methylation. HMGA proteins were coordinated or associated with histone and DNA modification and the expression of the factors related to pluripotency. CSCs showed remarkable differences compared with ESCs and iPSCs.
Collapse
Affiliation(s)
- Vincenzo Giancotti
- Department of Life Science, University of Trieste, Trieste, Italy
- Trieste Proteine Ricerche, Palmanova, Udine, Italy
| | - Natascha Bergamin
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Palmina Cataldi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Claudio Rizzi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| |
Collapse
|
26
|
Majidinia M, Aghazadeh J, Jahanban‐Esfahlani R, Yousefi B. The roles of Wnt/β‐catenin pathway in tissue development and regenerative medicine. J Cell Physiol 2018; 233:5598-5612. [DOI: 10.1002/jcp.26265] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/14/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Maryam Majidinia
- Solid Tumor Research CenterUrmia University of Medical SciencesUrmiaIran
| | - Javad Aghazadeh
- Department of NeurosurgeryUrmia University of Medical SciencesUrmiaIran
| | - Rana Jahanban‐Esfahlani
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
| | - Bahman Yousefi
- Stem Cell and Regenerative Medicine InstituteTabriz University of Medical SciencesTabrizIran
- Molecular Targeting Therapy Research GroupFaculty of MedicineTabriz University ofMedical SciencesTabrizIran
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Cardiometabolic diseases increasingly afflict our aging, dysmetabolic population. Complex signals regulating low-density lipoprotein receptor-related protein (LRP) and frizzled protein family members - the plasma membrane receptors for the cadre of Wnt polypeptide morphogens - contribute to the control of cardiovascular homeostasis. RECENT FINDINGS Both canonical (β-catenin-dependent) and noncanonical (β-catenin-independent) Wnt signaling programs control vascular smooth muscle (VSM) cell phenotypic modulation in cardiometabolic disease. LRP6 limits VSM proliferation, reduces arteriosclerotic transcriptional reprogramming, and preserves insulin sensitivity while LRP5 restrains foam cell formation. Adipose, skeletal muscle, macrophages, and VSM have emerged as important sources of circulating Wnt ligands that are dynamically regulated during the prediabetes-diabetes transition with cardiometabolic consequences. Platelets release Dkk1, a LRP5/LRP6 inhibitor that induces endothelial inflammation and the prosclerotic endothelial-mesenchymal transition. By contrast, inhibitory secreted frizzled-related proteins shape the Wnt signaling milieu to limit myocardial inflammation with ischemia-reperfusion injury. VSM sclerostin, an inhibitor of canonical Wnt signaling in bone, restrains remodeling that predisposes to aneurysm formation, and is downregulated in aneurysmal vessels by epigenetic methylation. SUMMARY Components of the Wnt signaling cascade represent novel targets for pharmacological intervention in cardiometabolic disease. Conversely, strategies targeting the Wnt signaling cascade for other therapeutic purposes will have cardiovascular consequences that must be delineated to establish clinically useful pharmacokinetic-pharmacodynamic relationships.
Collapse
Affiliation(s)
- Austin Gay
- Department of Internal Medicine-Endocrine Division, UT Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
28
|
Jin L, Cao Y, Yu G, Wang J, Lin X, Ge L, Du J, Wang L, Diao S, Lian X, Wang S, Dong R, Shan Z. SFRP2 enhances the osteogenic differentiation of apical papilla stem cells by antagonizing the canonical WNT pathway. Cell Mol Biol Lett 2017; 22:14. [PMID: 28794794 PMCID: PMC5547503 DOI: 10.1186/s11658-017-0044-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 07/17/2017] [Indexed: 01/26/2023] Open
Abstract
Background Exploring the molecular mechanisms underlying directed differentiation is helpful in the development of clinical applications of mesenchymal stem cells (MSCs). Our previous study on dental tissue-derived MSCs demonstrated that secreted frizzled-related protein 2 (SFRP2), a Wnt inhibitor, could enhance osteogenic differentiation in stem cells from the apical papilla (SCAPs). However, how SFRP2 promotes osteogenic differentiation of dental tissue-derived MSCs remains unclear. In this study, we used SCAPs to investigate the underlying mechanisms. Methods SCAPs were isolated from the apical papilla of immature third molars. Western blot and real-time RT-PCR were applied to detect the expression of β-catenin and Wnt target genes. Alizarin Red staining, quantitative calcium analysis, transwell cultures and in vivo transplantation experiments were used to study the osteogenic differentiation potential of SCAPs. Results SFRP2 inhibited canonical Wnt signaling by enhancing phosphorylation and decreasing the expression of nuclear β-catenin in vitro and in vivo. In addition, the target genes of the Wnt signaling pathway, AXIN2 (axin-related protein 2) and MMP7 (matrix metalloproteinase-7), were downregulated by SFRP2. WNT1 inhibited the osteogenic differentiation potential of SCAPs. SFRP2 could rescue this WNT1-impaired osteogenic differentiation potential. Conclusions The results suggest that SFRP2 could bind to locally present Wnt ligands and alter the balance of intracellular Wnt signaling to antagonize the canonical Wnt pathway in SCAPs. This elucidates the molecular mechanism underlying the SFRP2-mediated directed differentiation of SCAPs and indicates potential target genes for improving dental tissue regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s11658-017-0044-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Luyuan Jin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Yu Cao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Guoxia Yu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Department of Stomatology, Beijing Children's Hospital, Capital Medical University, No.56 Nanlishi Road, Xicheng District, Beijing, 100045 China
| | - Jinsong Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, No. 10 Xitoutiao Youanmen, Fengtai District, Beijing, 100069 China
| | - Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Department of Implant Dentistry, Capital Medical University School of Stomatology, Beijing, 100050 China
| | - Lihua Ge
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Juan Du
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Xiaomeng Lian
- Department of Stomatology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100045 China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, No. 10 Xitoutiao Youanmen, Fengtai District, Beijing, 100069 China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| | - Zhaochen Shan
- Oral and Maxillofacial Surgery Department, Capital Medical University School of Stomatology, No. 4 Tiantanxili, Dongcheng District, Beijing, 100050 China
| |
Collapse
|
29
|
|
30
|
Lin X, Dong R, Diao S, Yu G, Wang L, Li J, Fan Z. SFRP2 enhanced the adipogenic and neuronal differentiation potentials of stem cells from apical papilla. Cell Biol Int 2017; 41:534-543. [PMID: 28244619 DOI: 10.1002/cbin.10757] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 02/24/2017] [Indexed: 01/04/2023]
Abstract
Dental tissue-derived mesenchymal stem cells (MSCs) are easily obtained and considered as a favorable cell source for tissue engineering, but the regulation of direct differentiation is unknown, which restricts their application. The present study investigated the effect of SFRP2, a Wnt signaling modulator, on MSC differentiation using stem cells from apical papilla (SCAPs). The cells were cultured in specific inducing medium for adipogenic, neurogenic, or chondrogenic differentiation. Over-expression of SFRP2 via retroviral infection enhanced the adipogenic and neurogenic differentiation of SCAPs. While inhibit of Wnt pathway by IWR1-endo could enhance the neurogenic differentiation potentials of SCAPs, similar with the function of SFRP2. In addition, over-expression of SFRP2 up-regulated the expression of stemness-related genes SOX2 and OCT4. Furthermore, SOX2 and OCT4 expression was significantly inhibited after lentiviral silencing of SFRP2 in SCAPs. Therefore, our results suggest that SFRP2 enhances the adipogenic and neurogenic differentiation potentials of SCAPs by up-regulating SOX2 and OCT4. Moreover, the effect of SFRP2 in neurogenic differentiation of SCAPs maybe also associated with Wnt inhibition. Our results provided useful information about the molecular mechanism underlying directed differentiation in dental tissue-derived MSCs.
Collapse
Affiliation(s)
- Xiao Lin
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Department of Implant Dentistry, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Rui Dong
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| | - Shu Diao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Department of Pediatric Dentistry, Capital Medical University School of Stomatology, Beijing, China
| | - Guoxia Yu
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China.,Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China.,Department of Stomatology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Liping Wang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| | - Jun Li
- Department of Implant Dentistry, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
31
|
Malliaras K, Vakrou S, Kapelios CJ, Nanas JN. Innate heart regeneration: endogenous cellular sources and exogenous therapeutic amplification. Expert Opin Biol Ther 2016; 16:1341-1352. [PMID: 27484198 DOI: 10.1080/14712598.2016.1218846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The -once viewed as heretical- concept of the adult mammalian heart as a dynamic organ capable of endogenous regeneration has recently gained traction. However, estimated rates of myocyte turnover vary wildly and the underlying mechanisms of cardiac plasticity remain controversial. It is still unclear whether the adult mammalian heart gives birth to new myocytes through proliferation of resident myocytes, through cardiomyogenic differentiation of endogenous progenitors or through both mechanisms. AREAS COVERED In this review, the authors discuss the cellular origins of postnatal mammalian cardiomyogenesis and touch upon therapeutic strategies that could potentially amplify innate cardiac regeneration. EXPERT OPINION The adult mammalian heart harbors a limited but detectable capacity for spontaneous endogenous regeneration. During normal aging, proliferation of pre-existing cardiomyocytes is the dominant mechanism for generation of new cardiomyocytes. Following myocardial injury, myocyte proliferation increases modestly, but differentiation of endogenous progenitor cells appears to also contribute to cardiomyogenesis (although agreement on the latter point is not universal). Since cardiomyocyte deficiency underlies almost all types of heart disease, development of therapeutic strategies that amplify endogenous regeneration to a clinically-meaningful degree is of utmost importance.
Collapse
Affiliation(s)
- Konstantinos Malliaras
- a 3rd Department of Cardiology , University of Athens School of Medicine , Athens , Greece
| | - Styliani Vakrou
- a 3rd Department of Cardiology , University of Athens School of Medicine , Athens , Greece
| | - Chris J Kapelios
- a 3rd Department of Cardiology , University of Athens School of Medicine , Athens , Greece
| | - John N Nanas
- a 3rd Department of Cardiology , University of Athens School of Medicine , Athens , Greece
| |
Collapse
|
32
|
Bastakoty D, Young PP. Wnt/β-catenin pathway in tissue injury: roles in pathology and therapeutic opportunities for regeneration. FASEB J 2016; 30:3271-3284. [PMID: 27335371 DOI: 10.1096/fj.201600502r] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2016] [Indexed: 12/19/2022]
Abstract
The Wnt/β-catenin pathway is an evolutionarily conserved set of signals with critical roles in embryonic and neonatal development across species. In mammals the pathway is quiescent in many organs. It is reactivated in response to injury and is reported to play complex and contrasting roles in promoting regeneration and fibrosis. We review the current understanding of the role of the Wnt/β-catenin pathway in injury of various mammalian organs and discuss the current advances and potential of Wnt inhibitory therapeutics toward promoting tissue regeneration and reducing fibrosis.-Bastakoty, D., Young, P. P. Wnt/β-catenin pathway in tissue injury: roles in pathology and therapeutic opportunities for regeneration.
Collapse
Affiliation(s)
- Dikshya Bastakoty
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; and
| | - Pampee P Young
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; and Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
33
|
Hodgkinson CP, Bareja A, Gomez JA, Dzau VJ. Emerging Concepts in Paracrine Mechanisms in Regenerative Cardiovascular Medicine and Biology. Circ Res 2016; 118:95-107. [PMID: 26837742 DOI: 10.1161/circresaha.115.305373] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the past decade, substantial evidence supports the paradigm that stem cells exert their reparative and regenerative effects, in large part, through the release of biologically active molecules acting in a paracrine fashion on resident cells. The data suggest the existence of a tissue microenvironment where stem cell factors influence cell survival, inflammation, angiogenesis, repair, and regeneration in a temporal and spatial manner.
Collapse
Affiliation(s)
- Conrad P Hodgkinson
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - Akshay Bareja
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - José A Gomez
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - Victor J Dzau
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC.
| |
Collapse
|
34
|
Bastakoty D, Saraswati S, Joshi P, Atkinson J, Feoktistov I, Liu J, Harris JL, Young PP. Temporary, Systemic Inhibition of the WNT/β-Catenin Pathway promotes Regenerative Cardiac Repair following Myocardial Infarct. ACTA ACUST UNITED AC 2016; 2. [PMID: 28042617 DOI: 10.16966/2472-6990.111] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS The WNT/β-catenin pathway is temporarily activated in the heart following myocardial infarction (MI). Despite data from genetic models indicating both positive and negative roles for the WNT pathway depending on the model used, the effect of therapeutic inhibition of WNT pathway on post-injury outcome and the cellular mediators involved are not completely understood. Using a newly available, small molecule, GNF-6231, which averts WNT pathway activation by blocking secretion of all WNT ligands, we sought to investigate whether therapeutic inhibition of the WNT pathway temporarily after infarct can mitigate post injury cardiac dysfunction and fibrosis and the cellular mechanisms responsible for the effects. METHODS AND RESULTS Pharmacologic inhibition of the WNT pathway by post-MI intravenous injection of GNF-6231 in C57Bl/6 mice significantly reduced the decline in cardiac function (Fractional Shortening at day 30: 38.71 ± 4.13% in GNF-6231 treated vs. 34.89 ± 4.86% in vehicle-treated), prevented adverse cardiac remodeling, and reduced infarct size (9.07 ± 3.98% vs. 17.18 ± 4.97%). WNT inhibition augmented proliferation of interstitial cells, particularly in the distal myocardium, inhibited apoptosis of cardiomyocytes, and reduced myofibroblast proliferation in the peri-infarct region. In vitro studies showed that WNT inhibition increased proliferation of Sca1+ cardiac progenitors, improved survival of cardiomyocytes, and inhibited collagen I synthesis by cardiac myofibroblasts. CONCLUSION Systemic, temporary pharmacologic inhibition of the WNT pathway using an orally bioavailable drug immediately following MI resulted in improved function, reduced adverse remodeling and reduced infarct size in mice. Therapeutic WNT inhibition affected multiple aspects of infarct repair: it promoted proliferation of cardiac progenitors and other interstitial cells, inhibited myofibroblast proliferation, improved cardiomyocyte survival, and reduced collagen I gene expression by myofibroblasts. Our data point to a promising role for WNT inhibitory therapeutics as a new class of drugs to drive post-MI repair and prevent heart failure.
Collapse
Affiliation(s)
- Dikshya Bastakoty
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sarika Saraswati
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Piyush Joshi
- Interdisciplinary Graduate Program, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James Atkinson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Igor Feoktistov
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jun Liu
- Genomics Institute of Novartis Research Foundation, San Diego, California, USA
| | - Jennifer L Harris
- Genomics Institute of Novartis Research Foundation, San Diego, California, USA
| | - Pampee P Young
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA; Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|