1
|
Burggren W, Dzialowski E, Tzschentke B. The avian embryo as a time-honoured animal model in developmental, biomedical and agricultural research. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230438. [PMID: 40010394 PMCID: PMC11864840 DOI: 10.1098/rstb.2023.0438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 02/28/2025] Open
Abstract
Avian embryos have been at the core of embryological, morphological, physiological and biochemical/molecular research, especially involving research in three primary areas: developmental, biomedical and agricultural research. As developmental models, the avian embryo-especially that of the chicken-has been the single most used embryo model, perhaps in part from the combination of large size, ease of access and prior knowledge base. Developmental research with avian embryos has included organ system studies of the heart, vasculature, lungs, kidneys, nervous system, etc., as well as integrated physiological processes including gas-exchange, acid-base and ion/water regulation. In terms of translational research, avian embryos have modelled vascular development, based on the easily accessible chorioallantoic membrane under the eggshell. This same respiratory organ has enabled toxicological studies of how pollutants affect vertebrate development. Investigation of the transition to pulmonary breathing and the associated emergence of respiratory control has also relied heavily upon the avian embryo. In addition to developmental and biomedical investigations, the avian embryo has been studied intensively due to the huge importance of domesticated birds as a food source. Consequently, the effects of environment (including temperature, humidity, noise levels and photoperiod) during incubation on subsequent post-hatch phenotype are being actively investigated.This article is part of the theme issue 'The biology of the avian respiratory system'.
Collapse
Affiliation(s)
- Warren Burggren
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX76203-5017, USA
| | - Edward Dzialowski
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX76203-5017, USA
| | - Barbara Tzschentke
- Institute for Agricultural and Urban Ecological Projects (IASP) at Humboldt-Universität zu Berlin, Berlin10115, Germany
| |
Collapse
|
2
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
3
|
Ahmad W, Saleh B, Qazi REM, Muneer R, Khan I, Khan M, Salim A. Direct differentiation of rat skin fibroblasts into cardiomyocytes. Exp Cell Res 2024; 435:113934. [PMID: 38237847 DOI: 10.1016/j.yexcr.2024.113934] [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: 07/23/2023] [Revised: 12/16/2023] [Accepted: 01/13/2024] [Indexed: 01/28/2024]
Abstract
Myocardial infarction (MI) is one of the major cardiovascular diseases caused by diminished supply of nutrients and oxygen to the heart due to obstruction of the coronary artery. Different treatment options are available for cardiac diseases, however, they do not completely repair the damage. Therefore, reprogramming terminally differentiated fibroblasts using transcription factors is a promising strategy to differentiate them into cardiac like cells in vitro and to increase functional cardiomyocytes and reduce fibrotic scar in vivo. In this study, skin fibroblasts were selected for reprogramming because they serve as a convenient source for the autologous cell therapy. Fibroblasts were isolated from skin of rat pups, propagated, and directly reprogrammed towards cardiac lineage. For reprogramming, two different approaches were adopted, i.e., cells were transfected with: (1) combination of cardiac transcription factors; GATA4, MEF2c, Nkx2.5 (GMN), and (2) combination of cardiac transcription factors; GATA4, MEF2c, Nkx2.5, and iPSC factors; Oct4, Klf4, Sox2 and cMyc (GMNO). After 72 h of transfection, cells were analyzed for the expression of cardiac markers at the mRNA and protein levels. For in vivo study, rat MI models were developed by ligating the left anterior descending coronary artery and the reprogrammed cells were transplanted in the infarcted heart. qPCR results showed that the reprogrammed cells exhibited significant upregulation of cardiac genes. Immunocytochemistry analysis further confirmed cardiomyogenic differentiation of the reprogrammed cells. For the assessment of cardiac function, animals were analyzed via echocardiography after 2 and 4 weeks of cell transplantation. Echocardiographic results showed that the hearts transplanted with the reprogrammed cells improved ejection fraction, fractional shortening, left ventricular internal systolic and diastolic dimensions, and end systolic and diastolic volumes. After 4 weeks of cell transplantation, heart tissues were harvested and processed for histology. The histological analysis showed that the reprogrammed cells improved wall thickness of left ventricle and reduced fibrosis significantly as compared to the control. It is concluded from the study that novel combination of cardiac transcription factors directly reprogrammed skin fibroblasts and differentiated them into cardiomyocytes. These differentiated cells showed cardiomyogenic characters in vitro, and reduced fibrosis and improved cardiac function in vivo. Furthermore, direct reprogramming of fibroblasts transfected with cardiac transcription factors showed better regeneration of the injured myocardium and improved cardiac function as compared to the indirect approach in which combination of cardiac and iPSC factors were used. The study after further optimization could be used as a better strategy for cell-based therapeutic approaches for cardiovascular diseases.
Collapse
Affiliation(s)
- Waqas Ahmad
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Bilal Saleh
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rida-E-Maria Qazi
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rabbia Muneer
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Irfan Khan
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Mohsin Khan
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Asmat Salim
- Stem Cell Research Laboratory, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
4
|
Hu WS, Chen JY, Liao WY, Chang CH, Chen TS. Regulation of ROS/inflammasome Axis is Essential for Cardiac Regeneration in Aging Rats Receiving Transplantation of Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:1393-1401. [PMID: 38031779 DOI: 10.2174/011574888x276612231121065203] [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: 08/26/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Aging is a biological and gradual deterioration of function in living organisms. Aging is one of the risk factors for heart disease. OBJECTIVE Although mesenchymal stem cell transplantation shows potential in heart disease treatment, the relationship between stem cell-based therapy and oxidative stress/inflammasome axis regulation remains unclear. This study hypothesized that intervention of stem cells showed protective effect on heart aging induced by D-galactose through regulation of oxidative stress/inflammasome axis. METHODS An aging animal model was designed to test the above hypothesis. Experimental animals were divided into three groups, including Sham, D-gal (aging rats induced by d-galactose), and D-gal+WJSC (aging rats receiving mesenchymal stem cells). RESULTS Compared to the Sham, the experimental results indicate that structural alteration (HE stain and Masson's Trichrome stain), oxidative stress elevation (increase of TBARS level, expression of gp-91 and suppression of Sirt-1 as well as SOD2), increase of aging marker p53, suppression of cardiogenesis marker Troponin T, and inflammasome related protein markers expression (NLRP3, caspase-1 and IL-1 beta) were significantly observed in D-gal. In contrast, all pathological pathways were significantly improved in D-gal+WJSC when compared to D-gal. In addition, migration of stem cells to aging heart tissues was observed in the D-gal+WJSC group. CONCLUSION These findings suggest that mesenchymal stem cell transplantation effectively ameliorates aging hearts through oxidative stress/inflammasome axis regulation. The results from this study provide clinical potential for stem cell-based therapy in the treatment of aging hearts.
Collapse
Affiliation(s)
- Wei-Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung, 40042, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Jing-Yi Chen
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
| | - Wei-Yu Liao
- Department of Life Science, National Taiwan Normal University, Taipei, 11677, Taiwan
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 40237, Taiwan
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
| | - Chin-Hsien Chang
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 40237, Taiwan
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Taoyuan, 33303, Taiwan
- College of Chinese Medicine, China Medical University, Taichung City, 40402, Taiwan
| | - Tung-Sheng Chen
- Graduate Program of Biotechnology and Pharmaceutical Industries, National Taiwan Normal University, Taipei, 11677, Taiwan
| |
Collapse
|
5
|
Ramos TAR, Urquiza-Zurich S, Kim SY, Gillette TG, Hill JA, Lavandero S, do Rêgo TG, Maracaja-Coutinho V. Single-cell transcriptional landscape of long non-coding RNAs orchestrating mouse heart development. Cell Death Dis 2023; 14:841. [PMID: 38110334 PMCID: PMC10728149 DOI: 10.1038/s41419-023-06296-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 10/18/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
Long non-coding RNAs (lncRNAs) comprise the most representative transcriptional units of the mammalian genome. They are associated with organ development linked with the emergence of cardiovascular diseases. We used bioinformatic approaches, machine learning algorithms, systems biology analyses, and statistical techniques to define co-expression modules linked to heart development and cardiovascular diseases. We also uncovered differentially expressed transcripts in subpopulations of cardiomyocytes. Finally, from this work, we were able to identify eight cardiac cell-types; several new coding, lncRNA, and pcRNA markers; two cardiomyocyte subpopulations at four different time points (ventricle E9.5, left ventricle E11.5, right ventricle E14.5 and left atrium P0) that harbored co-expressed gene modules enriched in mitochondrial, heart development and cardiovascular diseases. Our results evidence the role of particular lncRNAs in heart development and highlight the usage of co-expression modular approaches in the cell-type functional definition.
Collapse
Grants
- R01 HL155765 NHLBI NIH HHS
- R01 HL126012 NHLBI NIH HHS
- R01 HL147933 NHLBI NIH HHS
- R01 HL128215 NHLBI NIH HHS
- R01 HL120732 NHLBI NIH HHS
- Agencia Nacional de Investigacion y Desarrollo (ANID, Chile), FONDAP 15130011 (SL), FONDECYT 1200490 (SL)
- the NIH: HL-120732 (JAH), HL-128215 (JAH), HL-126012 (JAH), HL-147933, (JAH), HL-155765 (JAH), 14SFRN20510023 (JAH), 14SFRN20670003 (JAH), Leducq grant number 11CVD04 (JAH), Cancer Prevention and Research Institute of Texas grant RP110486P3 (JAH)
- Agencia Nacional de Investigacion y Desarrollo (ANID, Chile), FONDAP 15130011 (VMC) and FONDECYT 1211731 (VMC).
Collapse
Affiliation(s)
- Thaís A R Ramos
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil
- Departamento de Informática, Centro de Informática, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Sebastián Urquiza-Zurich
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago, Chile.
| | - Thaís G do Rêgo
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil.
- Departamento de Informática, Centro de Informática, Universidade Federal da Paraíba, João Pessoa, Brazil.
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.
- Programa de Pós-Graduação em Bioinformática, Bioinformatics Multidisciplinary Environment (BioME), Instituto Metrópole Digital, Universidade Federal do Rio Grande do Norte, João Pessoa, Brazil.
| |
Collapse
|
6
|
The Human Induced Pluripotent Stem Cell Test as an Alternative Method for Embryotoxicity Testing. Int J Mol Sci 2022; 23:ijms23063295. [PMID: 35328717 PMCID: PMC8950674 DOI: 10.3390/ijms23063295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/01/2023] Open
Abstract
The evaluation of substances for their potency to induce embryotoxicity is controlled by safety regulations. Test guidelines for reproductive and developmental toxicity rely mainly on animal studies, which make up the majority of animal usage in regulatory toxicology. Therefore, there is an urgent need for alternative in vitro methods to follow the 3R principles. To improve human safety, cell models based on human cells are of great interest to overcome species differences. Here, human induced pluripotent stem cells (hiPSCs) are an ideal cell source as they largely recapitulate embryonic stem cells without bearing ethical concerns and they are able to differentiate into most cell types of the human body. Here, we set up and characterized a fetal bovine serum (FBS)-free hiPSC-based in vitro test method, called the human induced pluripotent stem cell test (hiPS Test), to evaluate the embryotoxic potential of substances. After 10 days in culture, hiPSCs develop into beating cardiomyocytes. As terminal endpoint evaluations, cell viability, qPCR analyses as well as beating frequency and area of beating cardiomyocytes by video analyses are measured. The embryotoxic positive and non-embryotoxic negative controls, 5-Fluorouracil (5-FU) and Penicillin G (PenG), respectively, were correctly assessed in the hiPS Test. More compounds need to be screened in the future for defining the assay’s applicability domain, which will inform us of the suitability of the hiPS Test for detecting adverse effects of substances on embryonic development.
Collapse
|
7
|
Katraki-Pavlou S, Kastana P, Bousis D, Ntenekou D, Varela A, Davos CH, Nikou S, Papadaki E, Tsigkas G, Athanasiadis E, Herradon G, Mikelis CM, Beis D, Papadimitriou E. Protein tyrosine phosphatase receptor zeta 1 deletion triggers defective heart morphogenesis in mice and zebrafish. Am J Physiol Heart Circ Physiol 2021; 322:H8-H24. [PMID: 34767486 PMCID: PMC8754060 DOI: 10.1152/ajpheart.00400.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein tyrosine phosphatase receptor-ζ1 (PTPRZ1) is a transmembrane
tyrosine phosphatase receptor highly expressed in embryonic stem cells. In the
present work, gene expression analyses of Ptprz1−/− and Ptprz1+/+ mice endothelial cells and hearts pointed to
an unidentified role of PTPRZ1 in heart development through the regulation of
heart-specific transcription factor genes. Echocardiography analysis in mice
identified that both systolic and diastolic functions are affected in Ptprz1−/− compared with Ptprz1+/+ hearts, based on a dilated left
ventricular (LV) cavity, decreased ejection fraction and fraction shortening,
and increased angiogenesis in Ptprz1−/−
hearts, with no signs of cardiac hypertrophy. A zebrafish ptprz1−/− knockout was also generated and exhibited
misregulated expression of developmental cardiac markers, bradycardia, and
defective heart morphogenesis characterized by enlarged ventricles and defected
contractility. A selective PTPRZ1 tyrosine phosphatase inhibitor affected
zebrafish heart development and function in a way like what is observed in the
ptprz1−/− zebrafish. The same
inhibitor had no effect in the function of the adult zebrafish heart, suggesting
that PTPRZ1 is not important for the adult heart function, in line with data
from the human cell atlas showing very low to negligible PTPRZ1 expression in
the adult human heart. However, in line with the animal models, Ptprz1 was expressed in many different cell types in
the human fetal heart, such as valvar, fibroblast-like, cardiomyocytes, and
endothelial cells. Collectively, these data suggest that PTPRZ1 regulates
cardiac morphogenesis in a way that subsequently affects heart function and
warrant further studies for the involvement of PTPRZ1 in idiopathic congenital
cardiac pathologies. NEW & NOTEWORTHY Protein tyrosine phosphatase receptor
ζ1 (PTPRZ1) is expressed in fetal but not adult heart and seems
to affect heart development. In both mouse and zebrafish animal models, loss of
PTPRZ1 results in dilated left ventricle cavity, decreased ejection fraction,
and fraction shortening, with no signs of cardiac hypertrophy. PTPRZ1 also seems
to be involved in atrioventricular canal specification, outflow tract
morphogenesis, and heart angiogenesis. These results suggest that PTPRZ1 plays a
role in heart development and support the hypothesis that it may be involved in
congenital cardiac pathologies.
Collapse
Affiliation(s)
- Stamatiki Katraki-Pavlou
- Zebrafish Disease Models Lab, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Greece.,Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Pinelopi Kastana
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Dimitris Bousis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Despoina Ntenekou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| | - Aimilia Varela
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Research Laboratory, Biomedical Research Foundation, Academy of Athens, Greece
| | - Sophia Nikou
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, Greece
| | - Eleni Papadaki
- Department of Anatomy-Histology-Embryology, Medical School, University of Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, Patras University Hospital, Rio, Patras, Greece
| | | | - Gonzalo Herradon
- Department of Pharmaceutical and Health Sciences, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Dimitris Beis
- Zebrafish Disease Models Lab, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Greece
| |
Collapse
|
8
|
Zhao A, Zhao K, Xia Y, Lyu J, Chen Y, Li S. Melatonin inhibits embryonic rat H9c2 cells growth through induction of apoptosis and cell cycle arrest via PI3K-AKT signaling pathway. Birth Defects Res 2021; 113:1171-1181. [PMID: 34231342 DOI: 10.1002/bdr2.1938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/10/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Our recent epidemiological study revealed that maternal sleep during the periconceptional period should be involved in the risk of congenital heart disease (CHD) in offspring. Melatonin, a sleep related hormone, has been suggested to play a crucial role in embryonic development based on the emerging evidence. In this study, we set out to assess the effect of melatonin on the embryonic cardiac cell growth and to explore the underlying mechanisms. METHODS We observed the effect of different gradient doses of melatonin as 10, 100, or 1,000 μM on cell proliferation in H9c2 embryonic rat cardiac cells. Furthermore, flow cytometry was applied to evaluate the impact on apoptosis and cell cycle. RNA-seq was conducted to screen the changes in expression of mRNA and signaling pathways. Quantitative Real-Time-PCR (qRT-PCR) was then conducted to validate the results. RESULTS It was observed that melatonin could inhibit H9c2 cell growth, at the doses of 100 and 1,000 μM, but not at 10 μM. Moreover, melatonin ranged from 100 to 1,000 μM could instigate cell cycle arrest at G1 phase and simulate apoptosis, in a dose-dependent manner. In addition, melatonin was found to down-regulate the expression of a number of genes, which are related to heart development (SPARC, IFITM3, TNNT2, LOX), and PI3K-Akt signaling pathway activation (FN1, HSP90B1, THBS1, MFGE8, and CLU). CONCLUSIONS Our findings suggested that high level of melatonin could be capable of inhibiting growth through the induction of apoptosis and cell cycle arrest via PI3K-AKT signaling pathway, thereby interfering with embryonic heart development. Considering this study is based on H9c2 embryonic rat cardiac cells, future additional studies using human embryonic cardiac cell are warranted.
Collapse
Affiliation(s)
- Anda Zhao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kena Zhao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanqing Xia
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Lyu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenghui Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Dakhel S, Davies WIL, Joseph JV, Tomar T, Remeseiro S, Gunhaga L. Chick fetal organ spheroids as a model to study development and disease. BMC Mol Cell Biol 2021; 22:37. [PMID: 34225662 PMCID: PMC8256237 DOI: 10.1186/s12860-021-00374-6] [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: 01/20/2021] [Accepted: 06/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Organ culture models have been used over the past few decades to study development and disease. The in vitro three-dimensional (3D) culture system of organoids is well known, however, these 3D systems are both costly and difficult to culture and maintain. As such, less expensive, faster and less complex methods to maintain 3D cell culture models would complement the use of organoids. Chick embryos have been used as a model to study human biology for centuries, with many fundamental discoveries as a result. These include cell type induction, cell competence, plasticity and contact inhibition, which indicates the relevance of using chick embryos when studying developmental biology and disease mechanisms. RESULTS Here, we present an updated protocol that enables time efficient, cost effective and long-term expansion of fetal organ spheroids (FOSs) from chick embryos. Utilizing this protocol, we generated FOSs in an anchorage-independent growth pattern from seven different organs, including brain, lung, heart, liver, stomach, intestine and epidermis. These three-dimensional (3D) structures recapitulate many cellular and structural aspects of their in vivo counterpart organs and serve as a useful developmental model. In addition, we show a functional application of FOSs to analyze cell-cell interaction and cell invasion patterns as observed in cancer. CONCLUSION The establishment of a broad ranging and highly effective method to generate FOSs from different organs was successful in terms of the formation of healthy, proliferating 3D organ spheroids that exhibited organ-like characteristics. Potential applications of chick FOSs are their use in studies of cell-to-cell contact, cell fusion and tumor invasion under defined conditions. Future studies will reveal whether chick FOSs also can be applicable in scientific areas such as viral infections, drug screening, cancer diagnostics and/or tissue engineering.
Collapse
Affiliation(s)
- Soran Dakhel
- Umeå Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
| | - Wayne I L Davies
- Umeå Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
| | - Justin V Joseph
- Umeå Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
| | - Tushar Tomar
- PamGene International B.V, Wolvenhoek 10, 5211 HH, 's-Hertogenbosch, The Netherlands
| | - Silvia Remeseiro
- Umeå Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden
| | - Lena Gunhaga
- Umeå Centre for Molecular Medicine, Umeå University, 901 87, Umeå, Sweden.
| |
Collapse
|
10
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
11
|
Genetic Restrictive Cardiomyopathy: Causes and Consequences-An Integrative Approach. Int J Mol Sci 2021; 22:ijms22020558. [PMID: 33429969 PMCID: PMC7827163 DOI: 10.3390/ijms22020558] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The sarcomere as the smallest contractile unit is prone to alterations in its functional, structural and associated proteins. Sarcomeric dysfunction leads to heart failure or cardiomyopathies like hypertrophic (HCM) or restrictive cardiomyopathy (RCM) etc. Genetic based RCM, a very rare but severe disease with a high mortality rate, might be induced by mutations in genes of non-sarcomeric, sarcomeric and sarcomere associated proteins. In this review, we discuss the functional effects in correlation to the phenotype and present an integrated model for the development of genetic RCM.
Collapse
|
12
|
Benslimane FM, Alser M, Zakaria ZZ, Sharma A, Abdelrahman HA, Yalcin HC. Adaptation of a Mice Doppler Echocardiography Platform to Measure Cardiac Flow Velocities for Embryonic Chicken and Adult Zebrafish. Front Bioeng Biotechnol 2019; 7:96. [PMID: 31139625 PMCID: PMC6527763 DOI: 10.3389/fbioe.2019.00096] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/16/2019] [Indexed: 02/06/2023] Open
Abstract
Ultrasonography is the most widely used imaging technique in cardiovascular medicine. In this technique, a piezoelectric crystal produces, sends, and receives high frequency ultrasound waves to the body to create an image of internal organs. It enables practical real time visualization in a non-invasive manner, making the modality especially useful to image dynamic cardiac structures. In the last few decades, echocardiography has been applied to in vivo cardiac disease models, mainly to rodents. While clinical echocardiography platforms can be used for relatively large animals such as pigs and rats, specialized systems are needed for smaller species. Theoretically, as the size of the imaged sample decreases, the frequency of the ultrasound transducer needed to image the sample increases. There are multiple modes of echocardiography imaging. In Doppler mode, erythrocytes blood flow velocities are measured from the frequency shift of the sent ultrasound waves compared to received echoes. Recorded data are then used to calculate cardiac function parameters such as cardiac output, as well as the hemodynamic shear stress levels in the heart and blood vessels. The multi-mode (i.e., b-mode, m-mode, Pulsed Doppler, Tissue Doppler, etc.) small animal ultrasound systems in the market can be used for most in vivo cardiac disease models including mice, embryonic chick and zebrafish. These systems are also associated with significant costs. Alternatively, there are more economical single-mode echocardiography platforms. However, these are originally built for mice studies and they need to be tested and evaluated for smaller experimental models. We recently adapted a mice Doppler echocardiography system to measure cardiac flow velocities for adult zebrafish and embryonic chicken. We successfully assessed cardiac function and hemodynamic shear stress for normal as well as for diseased embryonic chicken and zebrafish. In this paper, we will present our detailed protocols for Doppler flow measurements and further cardiac function analysis on these models using the setup. The protocols will involve detailed steps for animal stabilization, probe orientation for specific measurements, data acquisition, and data analysis. We believe this information will help cardiac researchers to establish similar echocardiography platforms in their labs in a practical and economical manner.
Collapse
Affiliation(s)
| | - Maha Alser
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Zain Z Zakaria
- Biomedical Research Center, Qatar University, Doha, Qatar.,Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Anju Sharma
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | | |
Collapse
|
13
|
Biswas A, Das S, Kapoor M, Shamsudheen KV, Jayarajan R, Verma A, Seth S, Bhargava B, Scaria V, Sivasubbu S, Rao V. Familial Hypertrophic Cardiomyopathy - Identification of cause and risk stratification through exome sequencing. Gene 2018; 660:151-156. [DOI: 10.1016/j.gene.2018.03.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/02/2018] [Accepted: 03/19/2018] [Indexed: 02/01/2023]
|
14
|
Mattapally S, Singh M, Murthy KS, Asthana S, Banerjee SK. Computational modeling suggests impaired interactions between NKX2.5 and GATA4 in individuals carrying a novel pathogenic D16N NKX2.5 mutation. Oncotarget 2018; 9:13713-13732. [PMID: 29568389 PMCID: PMC5862610 DOI: 10.18632/oncotarget.24459] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
NKX2.5, a homeobox containing gene, plays an important role in embryonic heart development and associated mutations are linked with various cardiac abnormalities. We sequenced the NKX2.5 gene in 100 congenital heart disease (CHD) patients and 200 controls. Our analysis revealed a total of 7 mutations, 3 in intronic region, 3 in coding region and 1 in 3’ UTR. Of the above mutations, one mutation was found to be associated with tetralogy of fallot (TOF) and two (rs2277923 and a novel mutation, D16N) were strongly associated with VSD. A novel missense mutation, D16N (p-value =0.009744), located in the tinman (TN) region and associated with ventricular septal defect (VSD), is the most significant findings of this study. Computational analysis revealed that D16N mutation is pathogenic in nature. Through the molecular modeling, docking and molecular dynamics simulation studies, we have identified the location of mutant D16N in NKX2.5 and its interaction map with other partners at the atomic level. We found NKX2.5-GATA4 complex is stable, however, in case of mutant we observed significant conformational changes and loss of key polar interactions, which might be a cause of the pathogenic behavior. This study underscores the structural basis of D16N pathogenic mutation in the regulation of NKX2.5 and how this mutation renders the structural-functional divergence that possibly leading towards the diseased state.
Collapse
Affiliation(s)
- Saidulu Mattapally
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mrityunjay Singh
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India
| | | | - Shailendra Asthana
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India
| | - Sanjay K Banerjee
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.,Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 121001, India
| |
Collapse
|
15
|
Pang KL, Parnall M, Loughna S. Effect of altered haemodynamics on the developing mitral valve in chick embryonic heart. J Mol Cell Cardiol 2017; 108:114-126. [PMID: 28576718 PMCID: PMC5529288 DOI: 10.1016/j.yjmcc.2017.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/23/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022]
Abstract
Intracardiac haemodynamics is crucial for normal cardiogenesis, with recent evidence showing valvulogenesis is haemodynamically dependent and inextricably linked with shear stress. Although valve anomalies have been associated with genetic mutations, often the cause is unknown. However, altered haemodynamics have been suggested as a pathogenic contributor to bicuspid aortic valve disease. Conversely, how abnormal haemodynamics impacts mitral valve development is still poorly understood. In order to analyse altered blood flow, the outflow tract of the chick heart was constricted using a ligature to increase cardiac pressure overload. Outflow tract-banding was performed at HH21, with harvesting at crucial valve development stages (HH26, HH29 and HH35). Although normal valve morphology was found in HH26 outflow tract banded hearts, smaller and dysmorphic mitral valve primordia were seen upon altered haemodynamics in histological and stereological analysis at HH29 and HH35. A decrease in apoptosis, and aberrant expression of a shear stress responsive gene and extracellular matrix markers in the endocardial cushions were seen in the chick HH29 outflow tract banded hearts. In addition, dysregulation of extracellular matrix (ECM) proteins fibrillin-2, type III collagen and tenascin were further demonstrated in more mature primordial mitral valve leaflets at HH35, with a concomitant decrease of ECM cross-linking enzyme, transglutaminase-2. These data provide compelling evidence that normal haemodynamics are a prerequisite for normal mitral valve morphogenesis, and abnormal blood flow could be a contributing factor in mitral valve defects, with differentiation as a possible underlying mechanism.
Collapse
Affiliation(s)
- Kar Lai Pang
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Matthew Parnall
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Siobhan Loughna
- School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
16
|
England J, Granados-Riveron J, Polo-Parada L, Kuriakose D, Moore C, Brook JD, Rutland CS, Setchfield K, Gell C, Ghosh TK, Bu'Lock F, Thornborough C, Ehler E, Loughna S. Tropomyosin 1: Multiple roles in the developing heart and in the formation of congenital heart defects. J Mol Cell Cardiol 2017; 106:1-13. [PMID: 28359939 PMCID: PMC5441184 DOI: 10.1016/j.yjmcc.2017.03.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/03/2022]
Abstract
Tropomyosin 1 (TPM1) is an essential sarcomeric component, stabilising the thin filament and facilitating actin's interaction with myosin. A number of sarcomeric proteins, such as alpha myosin heavy chain, play crucial roles in cardiac development. Mutations in these genes have been linked to congenital heart defects (CHDs), occurring in approximately 1 in 145 live births. To date, TPM1 has not been associated with isolated CHDs. Analysis of 380 CHD cases revealed three novel mutations in the TPM1 gene; IVS1 + 2T > C, I130V, S229F and a polyadenylation signal site variant GATAAA/AATAAA. Analysis of IVS1 + 2T > C revealed aberrant pre-mRNA splicing. In addition, abnormal structural properties were found in hearts transfected with TPM1 carrying I130V and S229F mutations. Phenotypic analysis of TPM1 morpholino-treated embryos revealed roles for TPM1 in cardiac looping, atrial septation and ventricular trabeculae formation and increased apoptosis was seen within the heart. In addition, sarcomere assembly was affected and altered action potentials were exhibited. This study demonstrated that sarcomeric TPM1 plays vital roles in cardiogenesis and is a suitable candidate gene for screening individuals with isolated CHDs. Four mutations identified in the TPM1 gene; IVS1 + 2T > C, I130V, S229F and GATAAA/AATAAA. In vitro analysis of IVS1 + 2T > C revealed aberrant pre-mRNA splicing. I130V and S229F mutations caused abnormal structural properties in the sarcomere. Reduced TPM1 expression during early cardiogenesis causes aberrant gross morphology. Apoptosis, sarcomere assembly and cardiac conduction were also affected.
Collapse
Affiliation(s)
| | - Javier Granados-Riveron
- Laboratory of Genomics, Genetics and Bioinformatics, Hospital Infantil de México Federico Gómez, Mexico
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, USA
| | | | | | - J David Brook
- School of Life Sciences, University of Nottingham, UK
| | - Catrin S Rutland
- School of Veterinary Medicine and Science, University of Nottingham, UK
| | | | | | | | - Frances Bu'Lock
- East Midlands Congenital Heart Centre, Glenfield Hospital, Leicester, UK
| | | | - Elisabeth Ehler
- Randall Division of Cell and Molecular Biophysics, The Cardiovascular Division, King's College London, UK
| | | |
Collapse
|