1
|
Zhao Y, Wan J, Liao B, Qi M. The Neglected Internodal Tract-A Cardiac Conduction System Structure Homologous to the Development and Regulation of the Sinoatrial Node. Rev Cardiovasc Med 2025; 26:27882. [PMID: 40351691 PMCID: PMC12059794 DOI: 10.31083/rcm27882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 05/14/2025] Open
Abstract
The existence of internodal tracts (ITs) is controversial. Indeed, ITs in the cardiac conduction system (CCS), connected to the sinoatrial node (SAN), transmit electrical signals quickly to the left atrium and the atrioventricular node (AVN). Interestingly, research has suggested that the ITs and the tail of the SAN may share developmental homology. Additionally, many studies indicate that ITs blockage can lead to atrial conduction block and is associated with atrial fibrillation (AF). However, few studies have been reported on the morphogenesis, development, and function of ITs. Therefore, this paper aims to review the morphogenesis, development, and function of ITs, focusing on the regulatory mechanisms of transcription factors (TFs), such as NK2 homeobox 5 (NKX2.5), SHOX homeobox 2 (SHOX2), hyperpolarization activated cyclic nucleotide gated potassium channel 4 (HCN4), and T-box transcription factor 3 (TBX3) in the development and morphogenesis of ITs. This review also explores the causes of arrhythmias, especially atrial block, in order to provide new insights into the pathogenesis of CCS disorders.
Collapse
Affiliation(s)
- Yuanqin Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital, Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Key Laboratory of Cardiovascular Remodeling and Dysfunction, 646000 Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Man Qi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Chinese People’s Liberation Army (PLA) General Hospital, College of Pulmonary & Critical Care Medicine, 100091 Beijing, China
- Department of Cardiology, Chinese PLA General Hospital, 100853 Beijing, China
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120 Shanghai, China
| |
Collapse
|
2
|
Basalamah F, Dilogo IH, Raharjo SB, Mansyur M, Siregar NC, Ibrahim N, Setianto BY, Yuniadi Y. TBX3 transfection and nodal signal pathway inhibition promote differentiation of adipose mesenchymal stem cell to cardiac pacemaker-like cells. Stem Cell Res Ther 2024; 15:148. [PMID: 38778426 PMCID: PMC11112768 DOI: 10.1186/s13287-024-03760-x] [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: 02/16/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are known as one of the best candidate cells to produce cardiac pacemaker-like cells (CPLCs). Upregulation of TBX3 transcription factor and inhibition of the nodal signal pathway have a significant role in the formation of cardiac pacemaker cells such as sinoatrial and atrioventricular nodes, which initiate the heartbeat and control the rhythm of heart contractions. This study aimed to confirm the effects of transfection of TBX3 transcription factor and inhibition of the nodal signal pathway on differentiating adipose-derived MSCs (AD-MSCs) to CPLCs. AD-MSCs were characterized using flow cytometry and three-lineage differentiation staining. METHODS The transfection of TBX3 plasmid was carried out using lipofectamine, and inhibition of the nodal signal pathway was done using the small-molecule SB431542. The morphology of the cells was observed using a light microscope. Pacemaker-specific markers, including TBX3, Cx30, HCN4, HCN1, HCN3, and KCNN4, were evaluated using the qRT-PCR method. For protein level, TBX3 and Cx30 were evaluated using ELISA and immunofluorescence staining. The electrophysiology of cells was evaluated using a patch clamp. RESULTS The TBX3 expression in the TBX3, SM, and TBX + SM groups significantly higher (p < 0.05) compared to the control group and cardiomyocytes. The expression of Cx40 and Cx43 genes were lower in TBX3, SM, TBX + SM groups. In contrast, Cx30 gene showed higher expression in TBX3 group. The expression HCN1, HCN3, and HCN4 genes are higher in TBX3 group. CONCLUSION The transfection of TBX3 and inhibition of the nodal signal pathway by small-molecule SB431542 enhanced differentiation of AD-MSCs to CPLCs.
Collapse
Affiliation(s)
- Faris Basalamah
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
- Faculty of Medicine and Health, University Muhammadiyah Jakarta, Banten, 15419, Indonesia
| | - Ismail Hadisoebroto Dilogo
- Department of Orthopedic and Traumatology, Faculty of Medicine, Universitas Indonesia-Dr Cipto Mangunkusumo National General Hospital, Jakarta, 10430, Indonesia
- Stem Cell and Tissue Engineering Research Cluster, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Dr Cipto Mangunkusumo National General Hospital, Jakarta, 10430, Indonesia
| | - Sunu Budhi Raharjo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia-National Heart Center Harapan Kita, Jakarta, 10430, Indonesia
| | - Muchtaruddin Mansyur
- Department of Community Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Nuryati Chairani Siregar
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Nurhadi Ibrahim
- Department of Medical Physiology and Biophysics, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
- Neuroscience and Brain Development Research Cluster, Indonesian Medical Education and Researsch Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Budi Yuli Setianto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Health and Nursing, Universitas Gajah Mada, Yogyakarta, 55281, Indonesia
| | - Yoga Yuniadi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia-National Heart Center Harapan Kita, Jakarta, 10430, Indonesia.
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia-RS Jantung Dan Pembuluh Darah Harapan Kita, Jakarta, 10420, Indonesia.
| |
Collapse
|
3
|
Li H, Tang Q, Yang T, Wang Z, Li D, Wang L, Li L, Chen Y, Huang H, Zhang Y, Chen Y. Segregation of morphogenetic regulatory function of Shox2 from its cell fate guardian role in sinoatrial node development. Commun Biol 2024; 7:385. [PMID: 38553636 PMCID: PMC10980793 DOI: 10.1038/s42003-024-06039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 03/11/2024] [Indexed: 04/02/2024] Open
Abstract
Shox2 plays a vital role in the morphogenesis and physiological function of the sinoatrial node (SAN), the primary cardiac pacemaker, manifested by the formation of a hypoplastic SAN and failed differentiation of pacemaker cells in Shox2 mutants. Shox2 and Nkx2-5 are co-expressed in the developing SAN and regulate the fate of the pacemaker cells through a Shox2-Nkx2-5 antagonistic mechanism. Here we show that simultaneous inactivation of Nkx2-5 in the SAN of Shox2 mutants (dKO) rescued the pacemaking cell fate but not the hypoplastic defects, indicating uncoupling of SAN cell fate determination and morphogenesis. Single-cell RNA-seq revealed that the presumptive SAN cells of Shox2-/- mutants failed to activate pacemaking program but remained in a progenitor state preceding working myocardium, while both wildtype and dKO SAN cells displayed normal pacemaking cell fate with similar cellular state. Shox2 thus acts as a safeguard but not a determinant to ensure the pacemaking cell fate through the Shox2-Nkx2-5 antagonistic mechanism, which is segregated from its morphogenetic regulatory function in SAN development.
Collapse
Affiliation(s)
- Hua Li
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fujian Medical University, Fuzhou, Fujian Province, 350122, PR China.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China.
| | - Qinghuang Tang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, 14214, USA
| | - Tianfang Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Zhengsen Wang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - Dainan Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Linyan Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, Sichuan Province, 610021, PR China
| | - Liwen Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Yaoyi Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Hai Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yanding Zhang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian Province, 350108, PR China
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
4
|
Li N, Zeng Y, Tai M, Lin B, Zhu D, Luo Y, Ren X, Zhu X, Li L, Wu H, Huang J. Analysis of the Prognostic Value and Gene Expression Mechanism of SHOX2 in Lung Adenocarcinoma. Front Mol Biosci 2021; 8:688274. [PMID: 34262939 PMCID: PMC8273341 DOI: 10.3389/fmolb.2021.688274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/26/2021] [Indexed: 01/15/2023] Open
Abstract
Background: Detection of SHOX2 methylation has been used to assist in the early diagnosis of lung cancer in many hospitals as SHOX2 may be important in the tumorigenesis of lung cancer. However, there are few studies on the mRNA expression, methylation, and molecular mechanism of SHOX2 in lung cancer. We aimed to explore the role of SHOX2 in lung adenocarcinoma (LUAD). Methods: First, we examined the differential expression of SHOX2 mRNA and methylation in cancerous and normal tissues using databases. Second, we analyzed the relationship between SHOX2 expression and common clinical parameters in LUAD patients. Third, we further explored the methylated level and its specific location of SHOX2 and the mainly factors of SHOX2 gene expression. Finally, we screened the correlatively expressed genes to analyze the pathways from the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes using DAVID. Results: We found that the mRNA expression of SHOX2 was higher in multiple cancers, including LUAD and lung squamous cell carcinoma (LUSC), than in normal tissues. Among LUAD patients, SHOX2 expression was higher in patients of middle–young age, with smoking history, in advanced stages, and with nodal distant metastasis. In addition, our results showed that patients with high expression of SHOX2 are prone to recurrence, poor differentiation, and poor prognosis. Thus, we identified that SHOX2 might be an oncogene for LUAD progression. The main factor influencing the high expression of SHOX2 mRNA may be DNA methylation, followed by copy number variation (CNV), but not by gene mutations in LUAD. Unexpectedly, we found that SHOX2 undergoes hypomethylation in the gene body instead of hypermethylation in the promoter. Additionally, SHOX2 has cross talk in the PI3K–Akt signaling pathway and ECM–receptor interaction. Conclusion:SHOX2 is highly expressed in most cancers. SHOX2 gene expression might be mainly regulated by methylation of its gene body in LUAD, and its high expression or hypomethylation indicates poor differentiation and poor prognosis. SHOX2 could be involved in PI3K–Akt and other important cancer-related signaling pathways to promote tumorigenesis.
Collapse
Affiliation(s)
- Nanhong Li
- Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yu Zeng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Min Tai
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Biyun Lin
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Di Zhu
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Yi Luo
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Xinle Ren
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Xiaoying Zhu
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Lanlan Li
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Hongrong Wu
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jian Huang
- The Center of Pathological Diagnosis and Research, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Department of Pathology, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
5
|
Hoffmann S, Roeth R, Diebold S, Gogel J, Hassel D, Just S, Rappold GA. Identification and Tissue-Specific Characterization of Novel SHOX-Regulated Genes in Zebrafish Highlights SOX Family Members Among Other Genes. Front Genet 2021; 12:688808. [PMID: 34122528 PMCID: PMC8191631 DOI: 10.3389/fgene.2021.688808] [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: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 02/01/2023] Open
Abstract
SHOX deficiency causes a spectrum of clinical phenotypes related to skeletal dysplasia and short stature, including Léri-Weill dyschondrosteosis, Langer mesomelic dysplasia, Turner syndrome, and idiopathic short stature. SHOX controls chondrocyte proliferation and differentiation, bone maturation, and cellular growth arrest and apoptosis via transcriptional regulation of its direct target genes NPPB, FGFR3, and CTGF. However, our understanding of SHOX-related pathways is still incomplete. To elucidate the underlying molecular mechanisms and to better understand the broad phenotypic spectrum of SHOX deficiency, we aimed to identify novel SHOX targets. We analyzed differentially expressed genes in SHOX-overexpressing human fibroblasts (NHDF), and confirmed the known SHOX target genes NPPB and FGFR among the most strongly regulated genes, together with 143 novel candidates. Altogether, 23 genes were selected for further validation, first by whole-body characterization in developing shox-deficient zebrafish embryos, followed by tissue-specific expression analysis in three shox-expressing zebrafish tissues: head (including brain, pharyngeal arches, eye, and olfactory epithelium), heart, and pectoral fins. Most genes were physiologically relevant in the pectoral fins, while only few genes were also significantly regulated in head and heart tissue. Interestingly, multiple sox family members (sox5, sox6, sox8, and sox18) were significantly dysregulated in shox-deficient pectoral fins together with other genes (nppa, nppc, cdkn1a, cdkn1ca, cyp26b1, and cy26c1), highlighting an important role for these genes in shox-related growth disorders. Network-based analysis integrating data from the Ingenuity pathways revealed that most of these genes act in a common network. Our results provide novel insights into the genetic pathways and molecular events leading to the clinical manifestation of SHOX deficiency.
Collapse
Affiliation(s)
- Sandra Hoffmann
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ralph Roeth
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany.,nCounter Core Facility, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Sabrina Diebold
- Clinic for Internal Medicine II - Molecular Cardiology, University Hospital Ulm, Ulm, Germany
| | - Jasmin Gogel
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - David Hassel
- Department of Internal Medicine III - Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Steffen Just
- Clinic for Internal Medicine II - Molecular Cardiology, University Hospital Ulm, Ulm, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| |
Collapse
|
6
|
Abstract
Cardiac development is a complex developmental process that is initiated soon after gastrulation, as two sets of precardiac mesodermal precursors are symmetrically located and subsequently fused at the embryonic midline forming the cardiac straight tube. Thereafter, the cardiac straight tube invariably bends to the right, configuring the first sign of morphological left–right asymmetry and soon thereafter the atrial and ventricular chambers are formed, expanded and progressively septated. As a consequence of all these morphogenetic processes, the fetal heart acquired a four-chambered structure having distinct inlet and outlet connections and a specialized conduction system capable of directing the electrical impulse within the fully formed heart. Over the last decades, our understanding of the morphogenetic, cellular, and molecular pathways involved in cardiac development has exponentially grown. Multiples aspects of the initial discoveries during heart formation has served as guiding tools to understand the etiology of cardiac congenital anomalies and adult cardiac pathology, as well as to enlighten novels approaches to heal the damaged heart. In this review we provide an overview of the complex cellular and molecular pathways driving heart morphogenesis and how those discoveries have provided new roads into the genetic, clinical and therapeutic management of the diseased hearts.
Collapse
|
7
|
Yechikov S, Kao HKJ, Chang CW, Pretto D, Zhang XD, Sun YH, Smithers R, Sirish P, Nolta JA, Chan JW, Chiamvimonvat N, Lieu DK. NODAL inhibition promotes differentiation of pacemaker-like cardiomyocytes from human induced pluripotent stem cells. Stem Cell Res 2020; 49:102043. [PMID: 33128951 PMCID: PMC7814970 DOI: 10.1016/j.scr.2020.102043] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/17/2020] [Accepted: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Directed cardiomyogenesis from human induced pluripotent stem cells (hiPSCs) has been greatly improved in the last decade but directed differentiation to pacemaking cardiomyocytes (CMs) remains incompletely understood. In this study, we demonstrated that inhibition of NODAL signaling by a specific NODAL inhibitor (SB431542) in the cardiac mesoderm differentiation stage downregulated PITX2c, a transcription factor that is known to inhibit the formation of the sinoatrial node in the left atrium during cardiac development. The resulting hiPSC-CMs were smaller in cell size, expressed higher pro-pacemaking transcription factors, TBX3 and TBX18, and exhibited pacemaking-like electrophysiological characteristics compared to control hiPSC-CMs differentiated from established Wnt-based protocol. The pacemaker-like subtype increased up to 2.4-fold in hiPSC-CMs differentiated with the addition of SB431542 relative to the control. Hence, Nodal inhibition in the cardiac mesoderm stage promoted pacemaker-like CM differentiation from hiPSCs. Improving the yield of human pacemaker-like CMs is a critical first step in the development of functional human cell-based biopacemakers.
Collapse
Affiliation(s)
- Sergey Yechikov
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Hillary K J Kao
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Che-Wei Chang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Dalyir Pretto
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Xiao-Dong Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA
| | - Yao-Hui Sun
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA
| | - Regan Smithers
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA; Bridges to Stem Cell Research Program, California State University Sacramento, Sacramento, CA 95819, USA
| | - Padmini Sirish
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Jan A Nolta
- Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA; Department of Hematology and Oncology, University of California, Davis, Sacramento, CA 95817, USA
| | - James W Chan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Department of Veterans Affairs, Northern California Health Care System, Mather, CA 95655, USA
| | - Deborah K Lieu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95817, USA; Institute for Regenerative Cures, University of California, Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
8
|
Martínez de Hoyo K, Ortega Enciso A, Mendoza Beltrán F, Reynolds Pombo J. Células madre como alternativa al marcapaso transvenoso. REVISTA COLOMBIANA DE CARDIOLOGÍA 2020. [DOI: 10.1016/j.rccar.2019.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
9
|
Li N, Zeng Y, Huang J. Signaling pathways and clinical application of RASSF1A and SHOX2 in lung cancer. J Cancer Res Clin Oncol 2020; 146:1379-1393. [PMID: 32266538 DOI: 10.1007/s00432-020-03188-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/17/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND An increasing number of studies have focused on the early diagnostic value of the methylation of RASSF1A and SHOX2 in lung cancer. However, the intricate cellular events related to RASSF1A and SHOX2 in lung cancer are still a mystery. For researchers and clinicians aiming to more profoundly understand the diagnostic value of methylated RASSF1A and SHOX2 in lung cancer, this review will provide deeper insights into the molecular events of RASSF1A and SHOX2 in lung cancer. METHODOLOGY We searched for relevant publications in the PubMed and Google Scholar databases using the keywords "RASSF1A", "SHOX2" and "lung cancer" etc. First, we reviewed the RASSF1A and SHOX2 genes, from their family structures to the functions of their basic structural domains. Then we mainly focused on the roles of RASSF1A and SHOX2 in lung cancer, especially on their molecular events in recent decades. Finally, we compared the value of measuring RASSF1A and SHOX2 gene methylation with that of the common methods for the diagnosis of lung cancer patients. RESULTS The RASSF1A and SHOX2 genes were confirmed to be regulators or effectors of multiple cancer signaling pathways, driving tumorigenesis and lung cancer progression. The detection of RASSF1A and SHOX2 gene methylation has higher sensitivity and specificity than other commonly used methods for diagnosing lung cancer, especially in the early stage. CONCLUSIONS The RASSF1A and SHOX2 genes are critical for the processes of tumorigenesis, development, metastasis, drug resistance, and recurrence in lung cancer. The combined detection of RASSF1A and SHOX2 gene methylation was identified as an excellent method for the screening and surveillance of lung cancer that exhibits high sensitivity and specificity.
Collapse
Affiliation(s)
- Nanhong Li
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yu Zeng
- Department of Respiration, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524003, China
| | - Jian Huang
- Department of Pathology, Guangdong Medical University, Zhanjiang, 524023, China.
- Pathological Diagnosis and Research Center, Affiliated Hospital, Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
10
|
Frasch MG, Giussani DA. Impact of Chronic Fetal Hypoxia and Inflammation on Cardiac Pacemaker Cell Development. Cells 2020; 9:E733. [PMID: 32192015 PMCID: PMC7140710 DOI: 10.3390/cells9030733] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic fetal hypoxia and infection are examples of adverse conditions during complicated pregnancy, which impact cardiac myogenesis and increase the lifetime risk of heart disease. However, the effects that chronic hypoxic or inflammatory environments exert on cardiac pacemaker cells are poorly understood. Here, we review the current evidence and novel avenues of bench-to-bed research in this field of perinatal cardiogenesis as well as its translational significance for early detection of future risk for cardiovascular disease.
Collapse
Affiliation(s)
- Martin G. Frasch
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA 98195, USA
- Center on Human Development and Disability, University of Washington, Seattle, WA 98195, USA
| | - Dino A. Giussani
- Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge CB2 1TN, UK;
| |
Collapse
|
11
|
Yin L, Liu MX, Wang FY, Wang X, Tang YH, Zhao QY, Wang T, Chen YT, Huang CX. Transcription Factor prrx1 Promotes Brown Adipose-Derived Stem Cells Differentiation to Sinus Node-Like Cells. DNA Cell Biol 2019; 38:1313-1322. [PMID: 31545082 DOI: 10.1089/dna.2019.4998] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study investigated whether overexpression of paired-related homeobox 1 (prrx1) can successfully induce differentiation of brown adipose-derived stem cells (BADSCs) into sinus node-like cells. The experiments were performed in two groups: adenovirus-green fluorescent protein (Ad-GFP) group and Ad-prrx1 group. After 5-7 days of adenoviral transfection, the expression levels of sinus node cell-associated pacing protein (hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 [HCN4]) and ion channel (calcium channel, voltage-dependent, T type, alpha 1G subunit [Cacna1g]), as well as transcription factors (T-box 18 [TBX18], insulin gene enhancer binding protein 1 [ISL-1], paired-like homeodomain transcription factor 2 [pitx2], short stature homeobox 2 [shox2]), were detected by western blot and reverse transcription-quantitative polymerase chain reaction. Immunofluorescence assay was carried out to detect whether prrx1 was coexpressed with HCN4, TBX18, and ISL-1. Finally, whole-cell patch-clamp technique was used to record pacing current hyperpolarization-activated inward current (If). The isolated cells were CD90+, CD29+, and CD45-, indicating that pure BADSCs were successfully isolated. After 5-7 days of Ad transfection into cells, the mRNA levels and protein levels of pacing-related factors (TBX18, ISL-1, HCN4, shox2, and Cacna1g) in Ad-prrx1 group were significantly higher than those in Ad-GFP group. However, the expression level of pitx2 was decreased. Immunofluorescence analysis showed that prrx1 was coexpressed with TBX18, ISL-1, and HCN4 in the Ad-prrx1 group, which did not appear in the Ad-GFP group. Whole-cell patch clamps were able to record the If current in the experimental group rather than in the Ad-GFP group. Overexpression of prrx1 can successfully induce sinus node-like cells.
Collapse
Affiliation(s)
- Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Ming-Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Feng-Yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Qing-Yan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Yu-Ting Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P.R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P.R. China
| |
Collapse
|
12
|
Feltes BC. Architects meets Repairers: The interplay between homeobox genes and DNA repair. DNA Repair (Amst) 2018; 73:34-48. [PMID: 30448208 DOI: 10.1016/j.dnarep.2018.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
Homeobox genes are widely considered the major protagonists of embryonic development and tissue formation. For the past decades, it was established that the deregulation of these genes is intimately related to developmental abnormalities and a broad range of diseases in adults. Since the proper regulation and expression of homeobox genes are necessary for a successful developmental program and tissue function, their relation to DNA repair mechanisms become a necessary discussion. However, important as it is, studies focused on the interplay between homeobox genes and DNA repair are scarce, and there is no critical discussion on the subject. Hence, in this work, I aim to provide the first review of the current knowledge of the interplay between homeobox genes and DNA repair mechanisms, and offer future perspectives on this, yet, young ground for new researches. Critical discussion is conducted, together with a careful assessment of each reviewed topic.
Collapse
Affiliation(s)
- Bruno César Feltes
- Institute of Informatics, Department of Theoretical Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|