1
|
Cao C, Li L, Zhang Q, Li H, Wang Z, Wang A, Liu J. Nkx2.5: a crucial regulator of cardiac development, regeneration and diseases. Front Cardiovasc Med 2023; 10:1270951. [PMID: 38124890 PMCID: PMC10732152 DOI: 10.3389/fcvm.2023.1270951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Cardiomyocytes fail to regenerate after birth and respond to mitotic signals through cellular hypertrophy rather than cellular proliferation. Necrotic cardiomyocytes in the infarcted ventricular tissue are eventually replaced by fibroblasts, generating scar tissue. Cardiomyocyte loss causes localized systolic dysfunction. Therefore, achieving the regeneration of cardiomyocytes is of great significance for cardiac function and development. Heart development is a complex biological process. An integral cardiac developmental network plays a decisive role in the regeneration of cardiomyocytes. During this process, genetic epigenetic factors, transcription factors, signaling pathways and small RNAs are involved in regulating the developmental process of the heart. Cardiomyocyte-specific genes largely promote myocardial regeneration, among which the Nkx2.5 transcription factor is one of the earliest markers of cardiac progenitor cells, and the loss or overexpression of Nkx2.5 affects cardiac development and is a promising candidate factor. Nkx2.5 affects the development and function of the heart through its multiple functional domains. However, until now, the specific mechanism of Nkx2.5 in cardiac development and regeneration is not been fully understood. Therefore, this article will review the molecular structure, function and interaction regulation of Nkx2.5 to provide a new direction for cardiac development and the treatment of heart regeneration.
Collapse
Affiliation(s)
- Ce Cao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Qian Zhang
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haoran Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ziyan Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Aoao Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
2
|
Cheung LYM, Menage L, Rizzoti K, Hamilton G, Dumontet T, Basham K, Daly AZ, Brinkmeier ML, Masser BE, Treier M, Cobb J, Delogu A, Lovell-Badge R, Hammer GD, Camper SA. Novel Candidate Regulators and Developmental Trajectory of Pituitary Thyrotropes. Endocrinology 2023; 164:bqad076. [PMID: 37183548 PMCID: PMC10227867 DOI: 10.1210/endocr/bqad076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations.
Collapse
Affiliation(s)
- Leonard Y M Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lucy Menage
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Greg Hamilton
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Typhanie Dumontet
- Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kaitlin Basham
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation: Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandre Z Daly
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Current affiliation is Vanguard, Valley Forge, PA 19482, USA
| | | | - Bailey E Masser
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mathias Treier
- Max Delbrϋck Center for Molecular Medicine (MDC), 13092 Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - John Cobb
- Department of Biological Sciences, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Alessio Delogu
- School of Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109, USA
- Endocrine Oncology Program, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Yin L, Wang FY, Zhang W, Wang X, Tang YH, Wang T, Chen YT, Huang CX. RA signaling pathway combined with Wnt signaling pathway regulates human-induced pluripotent stem cells (hiPSCs) differentiation to sinus node-like cells. Stem Cell Res Ther 2022; 13:324. [PMID: 35851424 PMCID: PMC9290266 DOI: 10.1186/s13287-022-03006-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The source of SAN is debated among researchers. Many studies have shown that RA and Wnt signaling are involved in heart development. In this study, we investigated the role of retinoic acid (RA) and Wnt signaling in the induction of sinus node-like cells. METHODS The experimental samples were divided into four groups: control group (CHIR = 0), CHIR = 3, RA + CHIR = 0 andRA + CHIR = 3. After 20 days of differentiation, Western blot, RT-qPCR, immunofluorescence and flow cytometry were performed to identify sinus node-like cells. Finally, whole-cell patch clamp technique was used to record pacing funny current and action potential (AP) in four groups. RESULTS The best intervention method used in our experiment was RA = 0.25 µmol/L D5-D9 + CHIR = 3 µmol/L D5-D7. Results showed that CHIR can increase the expression of ISL-1 and TBX3, while RA mainly elevated Shox2. Immunofluorescence assay and flow cytometry further illustrated that combining RA with CHIR can induce sinus node-like cells (CTNT+Shox2+Nkx2.5-). Moreover, CHIR might reduce the frequency of cell beats, but in conjunction with RA could partly compensate for this side effect. Whole cell patch clamps were able to record funny current and the typical sinus node AP in the experimental group, which did not appear in the control group. CONCLUSIONS Combining RA with Wnt signaling within a specific period can induce sinus node-like cells.
Collapse
Affiliation(s)
- Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Feng-yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Yan-hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Yu-ting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| | - Cong-xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060 Hubei People’s Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060 People’s Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060 People’s Republic of China
| |
Collapse
|
4
|
Yan Y, Liu F, Dang X, Zhou R, Liao B. TBX3 induces biased differentiation of human induced pluripotent stem cells into cardiac pacemaker-like cells. Gene Expr Patterns 2021; 40:119184. [PMID: 33975000 DOI: 10.1016/j.gep.2021.119184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/08/2021] [Accepted: 05/04/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND TBX3 plays a critical role in the formation of the sinoatrial node (SAN) during embryonic heart development. However, the contribution of TBX3 in driving the differentiation of human induced pluripotent stem cells (hiPSC)into pacemaker cells remains to be explored. RESULTS Using the pan-cardiomyocyte differentiation protocol of human induced pluripotent stem cells (hiPSC),TBX3 gene was introduced into the differentiating hiPSC on day 5 post-differentiation, and the differentiation of pacemaker-like cardiomyocytes was evaluated on day 21. The results showed that TBX3 significantly induced biased differentiation of hiPSC into pacemaker-like cells as judged by significantly increased expression of SAN-specific marker gene, SHOX2, and slightly decreased expression of SAN-detrimental transcription factor, NKX2-5. CONCLUSION Our results suggest that TBX3 plays an important role in driving the differentiation of hiPSC into pacemaker-like cells, and manipulation of TBX3 expression during pan-cardiomyocyte differentiation may lead to the development of therapeutic pacemaker cells.
Collapse
Affiliation(s)
- Ying Yan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China; College of Integrated Traditional Chinese and Western Medicine, Southwest Medical university, Luzhou, Sichuan, 646000, China.
| | - Feng Liu
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Xitong Dang
- The Key laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Rui Zhou
- The Key laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
5
|
Yu D, Febbo IG, Maroteaux MJ, Wang H, Song Y, Han X, Sun C, Meyer EE, Rowe S, Chen Y, Canavier CC, Schrader LA. The Transcription Factor Shox2 Shapes Neuron Firing Properties and Suppresses Seizures by Regulation of Key Ion Channels in Thalamocortical Neurons. Cereb Cortex 2021; 31:3194-3212. [PMID: 33675359 DOI: 10.1093/cercor/bhaa414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 01/02/2023] Open
Abstract
Thalamocortical neurons (TCNs) play a critical role in the maintenance of thalamocortical oscillations, dysregulation of which can result in certain types of seizures. Precise control over firing rates of TCNs is foundational to these oscillations, yet the transcriptional mechanisms that constrain these firing rates remain elusive. We hypothesized that Shox2 is a transcriptional regulator of ion channels important for TCN function and that loss of Shox2 alters firing frequency and activity, ultimately perturbing thalamocortical oscillations into an epilepsy-prone state. In this study, we used RNA sequencing and quantitative PCR of control and Shox2 knockout mice to determine Shox2-affected genes and revealed a network of ion channel genes important for neuronal firing properties. Protein regulation was confirmed by Western blotting, and electrophysiological recordings showed that Shox2 KO impacted the firing properties of a subpopulation of TCNs. Computational modeling showed that disruption of these conductances in a manner similar to Shox2's effects modulated frequency of oscillations and could convert sleep spindles to near spike and wave activity, which are a hallmark for absence epilepsy. Finally, Shox2 KO mice were more susceptible to pilocarpine-induced seizures. Overall, these results reveal Shox2 as a transcription factor important for TCN function in adult mouse thalamus.
Collapse
Affiliation(s)
- Diankun Yu
- Neuroscience Program, Brain Institute, Tulane University, USA
| | | | | | - Hanyun Wang
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Yingnan Song
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Xiao Han
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Cheng Sun
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Emily E Meyer
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Stuart Rowe
- Neuroscience Program, Brain Institute, Tulane University, USA
| | - Yiping Chen
- Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Carmen C Canavier
- Cell Biology and Anatomy, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Laura A Schrader
- Neuroscience Program, Brain Institute, Tulane University, USA.,Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
6
|
Conjugated activation of myocardial-specific transcription of Gja5 by a pair of Nkx2-5-Shox2 co-responsive elements. Dev Biol 2020; 465:79-87. [PMID: 32687896 DOI: 10.1016/j.ydbio.2020.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 02/08/2023]
Abstract
The sinoatrial node (SAN) is the primary pacemaker in the heart. During cardiogenesis, Shox2 and Nkx2-5 are co-expressed in the junction domain of the SAN and regulate pacemaker cell fate through a Shox2-Nkx2-5 antagonism. Cx40 is a marker of working myocardium and an Nkx2-5 transcriptional output antagonized by Shox2, but the underlying regulatory mechanisms remain elusive. Here we characterized a bona fide myocardial-specific Gja5 (coding gene of Cx40) distal enhancer consisting of a pair of Nkx2-5 and Shox2 co-bound elements in the regulatory region of Gja5. Transgenic reporter assays revealed that neither element alone, but the conjugation of both elements together, drives myocardial-specific transcription. Genetic analyses confirmed that the activation of this enhancer depends on Nkx2-5 but is inhibited by Shox2 in vivo, and its presence is essential for Gja5 expression in the myocardium but not the endothelial cells of the heart. Furthermore, chromatin conformation analysis showed an Nkx2-5-dependent loop formation between these two elements and the Gja5 promoter in vivo, indicating that Nkx2-5 bridges the conjugated activation of this enhancer by pairing the two elements to the Gja5 promoter.
Collapse
|
7
|
Li N, Wang ZS, Wang XH, Xu YJ, Qiao Q, Li XM, Di RM, Guo XJ, Li RG, Zhang M, Qiu XB, Yang YQ. A SHOX2 loss-of-function mutation underlying familial atrial fibrillation. Int J Med Sci 2018; 15:1564-1572. [PMID: 30443179 PMCID: PMC6216059 DOI: 10.7150/ijms.27424] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/29/2018] [Indexed: 01/01/2023] Open
Abstract
Atrial fibrillation (AF), as the most common sustained cardiac arrhythmia, is associated with substantially increased morbidity and mortality. Aggregating evidence demonstrates that genetic defects play a crucial role in the pathogenesis of AF, especially in familial AF. Nevertheless, AF is of pronounced genetic heterogeneity, and in an overwhelming majority of cases the genetic determinants underlying AF remain elusive. In the current study, 162 unrelated patients with familial AF and 238 unrelated healthy individuals served as controls were recruited. The coding exons and splicing junction sites of the SHOX2 gene, which encodes a homeobox-containing transcription factor essential for proper development and function of the cardiac conduction system, were sequenced in all study participants. The functional effect of the mutant SHOX2 protein was characterized with a dual-luciferase reporter assay system. As a result, a novel heterozygous SHOX2 mutation, c.580C>T or p.R194X, was identified in an index patient, which was absent from the 476 control chromosomes. Genetic analysis of the proband's pedigree revealed that the nonsense mutation co-segregated with AF in the family with complete penetrance. Functional assays demonstrated that the mutant SHOX2 protein had no transcriptional activity compared with its wild-type counterpart. In conclusion, this is the first report on the association of SHOX2 loss-of-function mutation with enhanced susceptibility to familial AF, which provides novel insight into the molecular mechanism underpinning AF, suggesting potential implications for genetic counseling and individualized management of AF patients.
Collapse
Affiliation(s)
- Ning Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Zhang-Sheng Wang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Xin-Hua Wang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai 200127, China
| | - Ying-Jia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Qi Qiao
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Xiu-Mei Li
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Ruo-Min Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Xiao-Juan Guo
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China.,Department of Cardiovascular Research Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Min Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 West Huaihai Road, Shanghai 200030, China
| | - Yi-Qing Yang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China.,Department of Cardiovascular Research Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China.,Department of Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai 200240, China
| |
Collapse
|
8
|
Zhou X, Hao Q, Liao P, Luo S, Zhang M, Hu G, Liu H, Zhang Y, Cao B, Baddoo M, Flemington EK, Zeng SX, Lu H. Nerve growth factor receptor negates the tumor suppressor p53 as a feedback regulator. eLife 2016; 5. [PMID: 27282385 PMCID: PMC4943851 DOI: 10.7554/elife.15099] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
Abstract
Cancer develops and progresses often by inactivating p53. Here, we unveil nerve growth factor receptor (NGFR, p75NTR or CD271) as a novel p53 inactivator. p53 activates NGFR transcription, whereas NGFR inactivates p53 by promoting its MDM2-mediated ubiquitin-dependent proteolysis and by directly binding to its central DNA binding domain and preventing its DNA-binding activity. Inversely, NGFR ablation activates p53, consequently inducing apoptosis, attenuating survival, and reducing clonogenic capability of cancer cells, as well as sensitizing human cancer cells to chemotherapeutic agents that induce p53 and suppressing mouse xenograft tumor growth. NGFR is highly expressed in human glioblastomas, and its gene is often amplified in breast cancers with wild type p53. Altogether, our results demonstrate that cancers hijack NGFR as an oncogenic inhibitor of p53. DOI:http://dx.doi.org/10.7554/eLife.15099.001 Cancer often develops as a result of alterations to “tumor suppressor” genes within cells. This results in the cells growing and dividing too much, which causes a tumor to form. One of the most important tumor suppressor genes produces a protein called p53, which is lost or mutated in roughly half of all human cancers. In the other half of cancers p53 itself is normal, but is often disabled by proteins that promote tumor growth. One of the remaining challenges in the field of cancer research is to identify which proteins inhibit p53 directly. Identifying these proteins would help clarify why many human cancers, such as some brain cancers, breast and skin cancers, often maintain a normal form of the p53 tumor suppressor protein. Zhou et al. now provide evidence that shows that a protein called nerve growth factor receptor (NGFR) is one such protein. NGFR was known to be important for the healthy development of the brain and nervous system. Unexpectedly, however, Zhou et al. found that NGFR binds directly to p53 and disables it in several types of human cancer cells. This finding is likely to be important because NGFR is produced in abnormally high amounts in several human cancer types, including skin, breast, bone and some brain cancers. Reducing the levels of NGFR in cancer cells caused the cells to become more sensitive to some anti-cancer drugs. Overall, the results presented by Zhou et al. suggest that developing new drugs that target NGFR could produce new treatments for human cancers that have a normal form of the gene that produces p53. More experiments are also needed to find out whether NGFR has other ways of promoting the development of cancerous tumors. DOI:http://dx.doi.org/10.7554/eLife.15099.002
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Qian Hao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Peng Liao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minhong Zhang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohui Hu
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongbing Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Yiwei Zhang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Bo Cao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Melody Baddoo
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Erik K Flemington
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Shelya X Zeng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| | - Hua Lu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, United States.,Tulane Cancer Center, Tulane University School of Medicine, New Orleans, United States
| |
Collapse
|
9
|
Feng Y, Yang P, Luo S, Zhang Z, Li H, Zhu P, Song Z. Shox2 influences mesenchymal stem cell fate in a co-culture model in vitro. Mol Med Rep 2016; 14:637-42. [PMID: 27222368 PMCID: PMC4918598 DOI: 10.3892/mmr.2016.5306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 02/24/2016] [Indexed: 01/26/2023] Open
Abstract
Sinoatrial node (SAN) dysfunction is a common cardiovascular problem, and the development of a cell sourced biological pacemaker has been the focus of cardiac electrophysiology research. The aim of biological pacemaker therapy is to produce SAN-like cells, which exhibit spontaneous activity characteristic of the SAN. Short stature homeobox 2 (Shox2) is an early cardiac transcription factor and is crucial in the formation and differentiation of the sinoatrial node (SAN). The present study aimed to improve pacemaker function by overexpression of Shox2 in canine mesenchymal stem cells (cMSCs) to induce a phenotype similar to native pacemaker cells. To achieve this objective, the cMSCs were transfected with lentiviral pLentis-mShox2-red fluorescent protein, and then co-cultured with rat neonatal cardiomyocytes (RNCMs) in vitro for 5–7 days. The feasibility of regulating the differentiation of cMSCs into pacemaker-like cells by Shox2 overexpression was investigated. Reverse transcription-quantitative polymerase chain reaction and western blotting showed that Shox2-transfected cMSCs expressed high levels of T box 3, hyperpolarization-activated cyclic nucleotide-gated cation channel and Connexin 45 genes, which participate in SAN development, and low levels of working myocardium genes, Nkx2.5 and Connexin 43. In addition, Shox2-transfected cMSCs were able to pace RNCMs with a rate faster than the control cells. In conclusion, these data indicate that overexpression of Shox2 in cMSCs can greatly enhance the pacemaker phenotype in a co-culture model in vitro.
Collapse
Affiliation(s)
- Yuanyuan Feng
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Pan Yang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shouming Luo
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhihui Zhang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Huakang Li
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ping Zhu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhiyuan Song
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
10
|
Heijman J, Heusch G, Dobrev D. Tools to Keep the Clock Ticking: Molecular Approaches to Treat Sinus Node Dysfunction. Cardiology 2015; 132:45-8. [DOI: 10.1159/000430783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 04/16/2015] [Indexed: 11/19/2022]
|