1
|
Liffers JL, Reinhardt JP, Seidl MD, Kirchhefer U, Müller FU, Schulte JS. Cre recombinase affects calcium dynamics already in young mice. Front Pharmacol 2025; 16:1558573. [PMID: 40206076 PMCID: PMC11979376 DOI: 10.3389/fphar.2025.1558573] [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: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Background The Cre/LoxP system is widely used in cardiovascular research to generate mouse models with tissue-specific inactivation of target genes. Studies have reported that expression of Cre recombinase under the αMHC promoter leads to age-dependent cardiotoxicity with ventricular hypertrophy, fibrosis and ventricular dysfunction at 6 months of age. This study explores the impact of Cre expression on intracellular Ca2+ dynamics in ventricular myocytes of αMHC-Cre mice as early as 3 months old. Methods Mice expressing Cre under the αMHC promoter (CRE) were compared to wild-type (WT) controls. Ventricular cardiomyocytes (VCMs) were isolated by the Langendorff method. Ca2+ transients and sarcomere shortening were simultaneously recorded from VCMs. Ventricular and atrial weights were assessed, VCM dimensions analyzed, and protein and mRNA levels of key proteins involved in Ca2+ dynamics measured by immunoblot analysis and quantitative real-time RT-PCR. Results At 3 months, CRE mice showed no evidence of cardiac hypertrophy. Ventricular or atrial weights and VCM size were not different between CRE and WT mice. The same applied to protein levels of SERCA2a, NCX1, Cav1.2, PLN and its phosphorylated form PLN pThr17. Nevertheless Ca2+ dynamics were significantly altered in CRE mice. Under basal conditions resting and peak Ca2+ were reduced and Ca2+ transient decay was delayed up to 30% in VCMs from CRE vs. WT mice. These differences persisted upon stimulation with 1 µM isoproterenol, whereas Ca2+ transient amplitude increased in CRE VCMs. We confirmed a previously reported reduction in dystrophin, potentially explaining the changes in Ca2+ dynamics. Despite these changes sarcomere shortening parameters were not different between groups. Conclusion As early as 3 months of age, Cre expression in VCMs leads to changes in Ca2+ dynamics that do not yet affect sarcomere shortening and cannot be attributed to the regulation of key proteins involved in Ca2+ dynamics. Because changes in intracellular Ca2+ dynamics can affect gene expression through altered excitation-transcription coupling, researchers should be aware of these subtle changes that precede the prominent phenotype at 6 months of age. Therefore, it is essential to use Cre-positive mice as controls when analyzing knockout models generated by the Cre/LoxP system.
Collapse
|
2
|
He K, Dong X, Yang T, Li Z, Liu Y, He J, Wu M, Wei-Zhang S, Kaysar P, Cui B, Yao X, Zhang L, Zhou W, Xu H, Wei J, Liu Q, Hu J, Wang X, Yan H. Smoking aggravates neovascular age-related macular degeneration via Sema4D-PlexinB1 axis-mediated activation of pericytes. Nat Commun 2025; 16:2821. [PMID: 40121188 PMCID: PMC11929803 DOI: 10.1038/s41467-025-58074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Age-related macular degeneration (AMD) is a prevalent neuroinflammation condition and the leading cause of irreversible blindness among the elderly population. Smoking significantly increases AMD risk, yet the mechanisms remain unclear. Here, we investigate the role of Sema4D-PlexinB1 axis in the progression of AMD, in which Sema4D-PlexinB1 is highly activated by smoking. Using patient-derived samples and mouse models, we discover that smoking increases the presence of Sema4D on the surface of CD8+ T cells that migrate into the choroidal neovascularization (CNV) lesion via CXCL12-CXCR4 axis and interact with its receptor PlexinB1 on choroidal pericytes. This leads to ROR2-mediated PlexinB1 phosphorylation and pericyte activation, thereby disrupting vascular homeostasis and promoting neovascularization. Inhibition of Sema4D reduces CNV and improves the benefit of anti-VEGF treatment. In conclusion, this study unveils the molecular mechanisms through which smoking exacerbates AMD pathology, and presents a potential therapeutic strategy by targeting Sema4D to augment current AMD treatments.
Collapse
Affiliation(s)
- Kai He
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, China
| | - Tianjing Yang
- School of Medicine, Nankai University, Tianjin, China
| | - Ziqi Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing He
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Meng Wu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, China
| | - Selena Wei-Zhang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Parhat Kaysar
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Bohao Cui
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueming Yao
- School of Medicine, Nankai University, Tianjin, China
| | - Li Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhou
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Jun Wei
- Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Junhao Hu
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| | - Xiaohong Wang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences; Tianjin Medical University, Tianjin, China.
| | - Hua Yan
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
3
|
Rahman SMN, Yung JHM, Volchuk A, Goldenberg NM, Giacca A. Metabolic phenotypes in a Lyz2Cre recombinase mouse model. Front Immunol 2025; 16:1499858. [PMID: 40170862 PMCID: PMC11958952 DOI: 10.3389/fimmu.2025.1499858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
The Cre-Lox system is essential in biomedical research for precise gene deletion in specific cell types, crucial for understanding genetic roles in disease. Although generally considered non-detrimental, Cre recombinase expression has been associated with potential adverse effects, including Cre toxicity, ectopic expression, and disruption of endogenous genes. We investigated the role of macrophage nucleotide-binding oligomerization domain (Nod1) in obesity-associated diabetes using myeloid-specific Nod1-knockout mice (Nod1 floxed crossed with Lyz2Cre). Our study examined Lyz2Cre as well as floxed control mice separately, unlike most research. Results indicated that Lyz2Cre expression alone impacts glucose metabolism, challenging the notion that Cre expression is harmless. This finding highlights the critical importance of including Cre-only controls in studies using floxed alleles to generate conditional knockout mouse models in order to ensure robust and accurate conclusions in molecular research.
Collapse
Affiliation(s)
- S. M. Niazur Rahman
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Justin Hou Ming Yung
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Allen Volchuk
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Neil M. Goldenberg
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Anesthesiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Zhao H, Zhou B. Lineage tracing of pancreatic cells for mechanistic and therapeutic insights. Trends Endocrinol Metab 2025:S1043-2760(24)00330-8. [PMID: 39828453 DOI: 10.1016/j.tem.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
Recent advances in lineage-tracing technologies have significantly improved our understanding of pancreatic cell biology, particularly in elucidating the ontogeny and regenerative capacity of pancreatic cells. A deeper appreciation of the mechanisms underlying pancreatic cell identity and plasticity holds the potential to inform the development of new therapeutic modalities for conditions such as diabetes and pancreatitis. With this goal in mind, here we summarize advances, challenges, and future directions in tracing pancreatic cell origins and fates using lineage-tracing technologies. Given their essential role for blood glucose regulation, we pay particular attention on the insights gained from endocrine cells, especially β-cells.
Collapse
Affiliation(s)
- Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
5
|
Co M, O’Brien GK, Wright KM, O’Roak BJ. Detailed phenotyping of Tbr1-2A-CreER knock-in mice demonstrates significant impacts on TBR1 protein levels and axon development. Autism Res 2024:10.1002/aur.3271. [PMID: 39548698 PMCID: PMC12078632 DOI: 10.1002/aur.3271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
Cre recombinase knock-in mouse lines have served as invaluable genetic tools for understanding key developmental processes altered in autism. However, insertion of exogenous DNA into the genome can have unintended effects on local gene regulation or protein function that must be carefully considered. Here, we analyze a recently generated Tbr1-2A-CreER knock-in mouse line, where a 2A-CreER cassette was inserted in-frame before the stop codon of the transcription factor gene Tbr1. Heterozygous TBR1 mutations in humans and mice are known to cause autism or autism-like behavioral phenotypes accompanied by structural brain malformations, most frequently a reduction of the anterior commissure (AC). Thus, it is critical for modified versions of Tbr1 to exhibit true wild-type-like activity. We evaluated the Tbr1-2A-CreER allele for its potential impact on Tbr1 function and complementation to Tbr1 loss-of-function alleles. In mice with one copy of the Tbr1-2A-CreER allele, we identified reduction of TBR1 protein in early postnatal cortex along with thinning of the AC, suggesting hypersensitivity of this structure to TBR1 dosage. Comparing Tbr1-2A-CreER and Tbr1-null mice to Tbr1-null complementation crosses showed reductions of TBR1 dosage ranging from 20% to 100%. Using six combinatorial genotypes, we found that moderate to severe TBR1 reductions (≥44%) were associated with cortical layer 5 expansion, while only the complete absence of TBR1 was associated with reeler-like "inverted" cortical layering. In total, these results strongly support the conclusion that Tbr1-2A-CreER is a hypomorphic allele. We advise caution when interpreting experiments using this allele, considering the sensitivity of various corticogenic processes to TBR1 dosage and the association of heterozygous TBR1 mutations with complex neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marissa Co
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Grace K. O’Brien
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Kevin M. Wright
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Brian J. O’Roak
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
6
|
Morikawa K, Nagasaki A, Sun L, Kawase E, Ebihara T, Shirayoshi Y. Optogenetic control of early embryos labeling using photoactivatable Cre recombinase 3.0. FEBS Open Bio 2024; 14:1888-1898. [PMID: 39223831 PMCID: PMC11532978 DOI: 10.1002/2211-5463.13862] [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: 11/04/2023] [Revised: 06/15/2024] [Accepted: 06/27/2024] [Indexed: 09/04/2024] Open
Abstract
Establishing a highly efficient photoactivatable Cre recombinase PA-Cre3.0 can allow spatiotemporal control of Cre recombinase activity. This technique may help to elucidate cell lineages, as well as facilitate gene and cell function analysis during development. This study examined the blue light-mediated optical regulation of Cre-loxP recombination using PA-Cre3.0 transgenic early mouse pre-implantation embryos. We found that inducing PA-Cre3.0 expression in the heterozygous state did not show detectable recombination activation with blue light. Conversely, in homozygous embryos, DNA recombination by PA-Cre3.0 was successfully induced by blue light and resulted in the activation of the red fluorescent protein reporter gene, while almost no leaks of Cre recombination activity were detected in embryos without light illumination. Thus, we characterize the conditions under which the PA-Cre3.0 system functions efficiently in early mouse embryos. These results are expected to provide a new optogenetic tool for certain biological studies, such as developmental process analysis and lineage tracing in early mouse embryos.
Collapse
Affiliation(s)
- Kumi Morikawa
- Cellular and Molecular Biotechnology Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Akira Nagasaki
- Biomedical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Lue Sun
- Health and Medical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Eihachiro Kawase
- Institute for Life and Medical SciencesKyoto UniversityKyotoJapan
| | - Tatsuhiko Ebihara
- Biomedical Research InstituteNational Institute of Advanced Industrial Science and Technology (AIST)TsukubaJapan
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Department of Genomic Medicine and Regenerative Therapy, Faculty of MedicineTottori UniversityYonagoJapan
| |
Collapse
|
7
|
Li BZ, Yuan YJ. Enhancing whole yeast genome rearrangements through multiple LoxPsym sequences. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2045-2047. [PMID: 38856790 DOI: 10.1007/s11427-024-2617-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Affiliation(s)
- Bing-Zhi Li
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
- Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China
| | - Ying-Jin Yuan
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China.
- Frontiers Research Institute for Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
8
|
Horvath C, Wolfrum C, Pelczar P. A safety guide for transgenic Cre drivers in metabolism. Nat Metab 2024; 6:1649-1652. [PMID: 39160335 DOI: 10.1038/s42255-024-01087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Affiliation(s)
- Carla Horvath
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Garcia-Gonzalez I, Rocha SF, Hamidi A, Garcia-Ortega L, Regano A, Sanchez-Muñoz M, Lytvyn M, Garcia-Cabero A, Roig-Soucase S, Schoofs H, Castro M, Sabata H, Potente M, Graupera M, Makinen T, Benedito R. iSuRe-HadCre is an essential tool for effective conditional genetics. Nucleic Acids Res 2024; 52:e56. [PMID: 38850155 PMCID: PMC11260470 DOI: 10.1093/nar/gkae472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/04/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024] Open
Abstract
Methods for modifying gene function at high spatiotemporal resolution in mice have revolutionized biomedical research, with Cre-loxP being the most widely used technology. However, the Cre-loxP technology has several drawbacks, including weak activity, leakiness, toxicity, and low reliability of existing Cre-reporters. This is mainly because different genes flanked by loxP sites (floxed) vary widely in their sensitivity to Cre-mediated recombination. Here, we report the generation, validation, and utility of iSuRe-HadCre, a new dual Cre-reporter and deleter mouse line that avoids these drawbacks. iSuRe-HadCre achieves this through a novel inducible dual-recombinase genetic cascade that ensures that cells expressing a fluorescent reporter had only transient Cre activity, that is nonetheless sufficient to effectively delete floxed genes. iSuRe-HadCre worked reliably in all cell types and for the 13 floxed genes tested. This new tool will enable the precise, efficient, and trustworthy analysis of gene function in entire mouse tissues or in single cells.
Collapse
Affiliation(s)
- Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Susana F Rocha
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Anahita Hamidi
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Lourdes Garcia-Ortega
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alvaro Regano
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Maria S Sanchez-Muñoz
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Mariya Lytvyn
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aroa Garcia-Cabero
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergi Roig-Soucase
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Hans Schoofs
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Marco Castro
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helena Sabata
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- ICREA, Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, Barcelona, Spain
| | - Taija Makinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
- Translational Cancer Medicine Program, Research Programs Unit, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
- Wihuri Research Institute, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
10
|
Tsisanova E, Nobles M, Sebastian S, Ng KE, Thomas A, Weinstein LS, Munroe PB, Tinker A. The ric-8b protein (resistance to inhibitors of cholinesterase 8b) is key to preserving contractile function in the adult heart. J Biol Chem 2024; 300:107470. [PMID: 38879012 PMCID: PMC11277413 DOI: 10.1016/j.jbc.2024.107470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 07/09/2024] Open
Abstract
Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function, but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis, and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodeling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the β-adrenergic pathways. Using fluorescence resonance energy transfer-based assays, we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.
Collapse
Affiliation(s)
- Elena Tsisanova
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muriel Nobles
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sonia Sebastian
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Keat-Eng Ng
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alison Thomas
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Patricia B Munroe
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrew Tinker
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
11
|
Dai P, Ma C, Chen C, Liang M, Dong S, Chen H, Zhang X. Unlocking Genetic Mysteries during the Epic Sperm Journey toward Fertilization: Further Expanding Cre Mouse Lines. Biomolecules 2024; 14:529. [PMID: 38785936 PMCID: PMC11117649 DOI: 10.3390/biom14050529] [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: 03/22/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
The spatiotemporal expression patterns of genes are crucial for maintaining normal physiological functions in animals. Conditional gene knockout using the cyclization recombination enzyme (Cre)/locus of crossover of P1 (Cre/LoxP) strategy has been extensively employed for functional assays at specific tissue or developmental stages. This approach aids in uncovering the associations between phenotypes and gene regulation while minimizing interference among distinct tissues. Various Cre-engineered mouse models have been utilized in the male reproductive system, including Dppa3-MERCre for primordial germ cells, Ddx4-Cre and Stra8-Cre for spermatogonia, Prm1-Cre and Acrv1-iCre for haploid spermatids, Cyp17a1-iCre for the Leydig cell, Sox9-Cre for the Sertoli cell, and Lcn5/8/9-Cre for differentiated segments of the epididymis. Notably, the specificity and functioning stage of Cre recombinases vary, and the efficiency of recombination driven by Cre depends on endogenous promoters with different sequences as well as the constructed Cre vectors, even when controlled by an identical promoter. Cre mouse models generated via traditional recombination or CRISPR/Cas9 also exhibit distinct knockout properties. This review focuses on Cre-engineered mouse models applied to the male reproductive system, including Cre-targeting strategies, mouse model screening, and practical challenges encountered, particularly with novel mouse strains over the past decade. It aims to provide valuable references for studies conducted on the male reproductive system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226001, China; (P.D.); (C.M.); (C.C.); (M.L.); (S.D.); (H.C.)
| |
Collapse
|
12
|
Klessinger D, Mamazhakypov A, Glaeser S, Emig R, Peyronnet R, Meier L, Proelss K, Marenne K, Smolka C, Grundmann S, Pankratz F, Esser PR, Moser M, Zhou Q, Esser JS. Divergent and Compensatory Effects of BMP2 and BMP4 on the VSMC Phenotype and BMP4's Role in Thoracic Aortic Aneurysm Development. Cells 2024; 13:735. [PMID: 38727271 PMCID: PMC11083443 DOI: 10.3390/cells13090735] [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: 04/05/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Angiotensin II/pharmacology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Apoptosis/drug effects
- Bone Morphogenetic Protein 2/metabolism
- Bone Morphogenetic Protein 4/metabolism
- Disease Models, Animal
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Signal Transduction
Collapse
Affiliation(s)
- Daniel Klessinger
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg im Breisgau, Germany;
| | - Sophie Glaeser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany; (R.E.); (R.P.)
- CIBSS Centre for Integrative Biological Signalling Studies, Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Remi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany; (R.E.); (R.P.)
| | - Lena Meier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Kora Proelss
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Katia Marenne
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Christian Smolka
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Sebastian Grundmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Franziska Pankratz
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Philipp R. Esser
- Allergy Research Group, Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany;
| | - Martin Moser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Qian Zhou
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Jennifer S. Esser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| |
Collapse
|
13
|
Cathcart B, Cheedipudi SM, Rouhi L, Zhao Z, Gurha P, Marian AJ. DNA double-stranded breaks, a hallmark of aging, defined at the nucleotide resolution, are increased and associated with transcription in the cardiac myocytes in LMNA-cardiomyopathy. Cardiovasc Res 2024:cvae063. [PMID: 38577741 DOI: 10.1093/cvr/cvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
AIMS An intrinsic feature of gene transcription is the formation of DNA superhelices near the transcription bubble, which are resolved upon induction of transient double-stranded breaks (DSBs) by topoisomerases. Unrepaired DSBs are pathogenic as they lead to cell cycle arrest, senescence, inflammation, and organ dysfunction. We posit that DSBs would be more prevalent at the genomic sites that are associated with gene expression. The objectives were to identify and characterize genome-wide DSBs at the nucleotide resolution and determine the association of DSBs with transcription in cardiac myocytes. METHODS AND RESULTS We identified the genome-wide DSBs in ∼1 million cardiac myocytes per heart in three wild-type and three myocyte-specific LMNA-deficient (Myh6-Cre:LmnaF/F) mice by END-Sequencing. The prevalence of DSBs was 0.8% and 2.2% in the wild-type and Myh6-Cre:LmnaF/F myocytes, respectively. The END-Seq signals were enriched for 8 and 6764 DSBs in the wild-type and Myh6-Cre:LmnaF/F myocytes, respectively (q < 0.05). The DSBs were preferentially localized to the gene regions, transcription initiation sites, cardiac transcription factor motifs, and the G quadruplex forming structures. Because LMNA regulates transcription through the lamin-associated domains (LADs), we defined the LADs in cardiac myocytes by a Cleavage Under Targets & Release Using Nuclease (CUT&RUN) assay (N = 5). On average there were 818 LADs per myocyte. Constitutive LADs (cLADs), defined as LADs that were shared by at least three genomes (N = 2572), comprised about a third of the mouse cardiac myocyte genomes. Transcript levels of the protein-coding genes located at the cLADs (N = 3975) were ∼16-fold lower than those at the non-LAD regions (N = ∼17 778). The prevalence of DSBs was higher in the non-LAD as compared to the cLAD regions. Likewise, DSBs were more common in the loss-of-LAD regions, defined as the genomic regions in the Myh6-Cre:LmnaF/F that were juxtaposed to the LAD regions in the wild-type myocytes. CONCLUSION To our knowledge, this is the first identification of the DSBs, at the nucleotide resolution in the cardiovascular system. The prevalence of DSBs was higher in the genomic regions associated with transcription. Because transcription is pervasive, DSBs are expected to be common and pathogenic in various states and aging.
Collapse
Affiliation(s)
- Benjamin Cathcart
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, UTHealth, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| |
Collapse
|
14
|
Li B, Bai WW, Guo T, Tang ZY, Jing XJ, Shan TC, Yin S, Li Y, Wang F, Zhu ML, Lu JX, Bai YP, Dong B, Li P, Wang SX. Statins improve cardiac endothelial function to prevent heart failure with preserved ejection fraction through upregulating circRNA-RBCK1. Nat Commun 2024; 15:2953. [PMID: 38580662 PMCID: PMC10997751 DOI: 10.1038/s41467-024-47327-z] [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: 11/03/2021] [Accepted: 03/27/2024] [Indexed: 04/07/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is associated with endothelial dysfunction. We have previously reported that statins prevent endothelial dysfunction through inhibition of microRNA-133a (miR-133a). This study is to investigate the effects and the underlying mechanisms of statins on HFpEF. Here, we show that statins upregulate the expression of a circular RNA (circRNA-RBCK1) which is co-transcripted with the ring-B-box-coiled-coil protein interacting with protein kinase C-1 (RBCK1) gene. Simultaneously, statins increase activator protein 2 alpha (AP-2α) transcriptional activity and the interaction between circRNA-RBCK1 and miR-133a. Furthermore, AP-2α directly interacts with RBCK1 gene promoter in endothelial cells. In vivo, lovastatin improves diastolic function in male mice under HFpEF, which is abolished by loss function of endothelial AP-2α or circRNA-RBCK1. This study suggests that statins upregulate the AP-2α/circRNA-RBCK1 signaling to suppress miR-133a in cardiac endothelial cells and prevent diastolic dysfunction in HFpEF.
Collapse
Affiliation(s)
- Bin Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wen-Wu Bai
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Guo
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhen-Yu Tang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xue-Jiao Jing
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ti-Chao Shan
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Sen Yin
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ying Li
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Fu Wang
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mo-Li Zhu
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jun-Xiu Lu
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yong-Ping Bai
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan, China
- Department of Geriatric Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bo Dong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Peng Li
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shuang-Xi Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
15
|
Asunción-Alvarez D, Palacios J, Ybañez-Julca RO, Rodriguez-Silva CN, Nwokocha C, Cifuentes F, Greensmith DJ. Calcium signaling in endothelial and vascular smooth muscle cells: sex differences and the influence of estrogens and androgens. Am J Physiol Heart Circ Physiol 2024; 326:H950-H970. [PMID: 38334967 DOI: 10.1152/ajpheart.00600.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Calcium signaling in vascular endothelial cells (ECs) and smooth muscle cells (VSMCs) is essential for the regulation of vascular tone. However, the changes to intracellular Ca2+ concentrations are often influenced by sex differences. Furthermore, a large body of evidence shows that sex hormone imbalance leads to dysregulation of Ca2+ signaling and this is a key factor in the pathogenesis of cardiovascular diseases. In this review, the effects of estrogens and androgens on vascular calcium-handling proteins are discussed, with emphasis on the associated genomic or nongenomic molecular mechanisms. The experimental models from which data were collected were also considered. The review highlights 1) in female ECs, transient receptor potential vanilloid 4 (TRPV4) and mitochondrial Ca2+ uniporter (MCU) enhance Ca2+-dependent nitric oxide (NO) generation. In males, only transient receptor potential canonical 3 (TRPC3) plays a fundamental role in this effect. 2) Female VSMCs have lower cytosolic Ca2+ levels than males due to differences in the activity and expression of stromal interaction molecule 1 (STIM1), calcium release-activated calcium modulator 1 (Orai1), calcium voltage-gated channel subunit-α1C (CaV1.2), Na+-K+-2Cl- symporter (NKCC1), and the Na+/K+-ATPase. 3) When compared with androgens, the influence of estrogens on Ca2+ homeostasis, vascular tone, and incidence of vascular disease is better documented. 4) Many studies use supraphysiological concentrations of sex hormones, which may limit the physiological relevance of outcomes. 5) Sex-dependent differences in Ca2+ signaling mean both sexes ought to be included in experimental design.
Collapse
Affiliation(s)
- Daniel Asunción-Alvarez
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Javier Palacios
- Laboratorio de Bioquímica Aplicada, Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Roberto O Ybañez-Julca
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Cristhian N Rodriguez-Silva
- Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Chukwuemeka Nwokocha
- Department of Basic Medical Sciences Physiology Section, Faculty of Medical Sciences, The University of the West Indies, Kingston, Jamaica
| | - Fredi Cifuentes
- Laboratorio de Fisiología Experimental (EphyL), Instituto Antofagasta (IA), Universidad de Antofagasta, Antofagasta, Chile
| | - David J Greensmith
- Biomedical Research Centre, School of Science, Engineering and Environment, The University of Salford, Salford, United Kingdom
| |
Collapse
|
16
|
Ding WX, Ma X, Kim S, Wang S, Ni HM. Recent insights about autophagy in pancreatitis. EGASTROENTEROLOGY 2024; 2:e100057. [PMID: 38770349 PMCID: PMC11104508 DOI: 10.1136/egastro-2023-100057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Acute pancreatitis is a common inflammatory gastrointestinal disease without any successful treatment. Pancreatic exocrine acinar cells have high rates of protein synthesis to produce and secrete large amounts of digestive enzymes. When the regulation of organelle and protein homeostasis is disrupted, it can lead to endoplasmic reticulum (ER) stress, damage to the mitochondria and improper intracellular trypsinogen activation, ultimately resulting in acinar cell damage and the onset of pancreatitis. To balance the homeostasis of organelles and adapt to protect themselves from organelle stress, cells use protective mechanisms such as autophagy. In the mouse pancreas, defective basal autophagy disrupts ER homoeostasis, leading to ER stress and trypsinogen activation, resulting in spontaneous pancreatitis. In this review, we discuss the regulation of autophagy and its physiological role in maintaining acinar cell homeostasis and function. We also summarise the current understanding of the mechanisms and the role of defective autophagy at multiple stages in experimental pancreatitis induced by cerulein or alcohol.
Collapse
Affiliation(s)
- Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sydney Kim
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
17
|
Mansouri M, Fussenegger M. Small-Molecule Regulators for Gene Switches to Program Mammalian Cell Behaviour. Chembiochem 2024; 25:e202300717. [PMID: 38081780 DOI: 10.1002/cbic.202300717] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Synthetic or natural small molecules have been extensively employed as trigger signals or inducers to regulate engineered gene circuits introduced into living cells in order to obtain desired outputs in a controlled and predictable manner. Here, we provide an overview of small molecules used to drive synthetic-biology-based gene circuits in mammalian cells, together with examples of applications at different levels of control, including regulation of DNA manipulation, RNA synthesis and editing, and protein synthesis, maturation, and trafficking. We also discuss the therapeutic potential of these small-molecule-responsive gene circuits, focusing on the advantages and disadvantages of using small molecules as triggers, the mechanisms involved, and the requirements for selecting suitable molecules, including efficiency, specificity, orthogonality, and safety. Finally, we explore potential future directions for translation of these devices to clinical medicine.
Collapse
Affiliation(s)
- Maysam Mansouri
- ETH Zurich, Department of Biosystems Science and Engineering, Klingelbergstrasse 48, CH-4056, Basel, Switzerland
| | - Martin Fussenegger
- ETH Zurich, Department of Biosystems Science and Engineering, Klingelbergstrasse 48, CH-4056, Basel, Switzerland
- University of Basel, Faculty of Science, Klingelbergstrasse 48, CH-4056, Basel, Switzerland
| |
Collapse
|
18
|
Man AWC, Zhou Y, Reifenberg G, Camp A, Münzel T, Daiber A, Xia N, Li H. Deletion of adipocyte NOS3 potentiates high-fat diet-induced hypertension and vascular remodelling via chemerin. Cardiovasc Res 2023; 119:2755-2769. [PMID: 37897505 PMCID: PMC10757584 DOI: 10.1093/cvr/cvad164] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/17/2023] [Accepted: 10/27/2023] [Indexed: 10/30/2023] Open
Abstract
AIMS Obesity is an epidemic that is a critical contributor to hypertension and other cardiovascular diseases. Current paradigms suggest that endothelial nitric oxide synthase (eNOS/NOS3) in the vessel wall is the primary regulator of vascular function and blood pressure. However, recent studies have revealed the presence of eNOS/NOS3 in the adipocytes of white adipose tissues and perivascular adipose tissues (PVATs). The current understanding of the role of adipocyte NOS3 is based mainly on studies using global knockout models. The present study aimed to elucidate the functional significance of adipocyte NOS3 for vascular function and blood pressure control. METHODS AND RESULTS We generated an adipocyte-specific NOS3 knockout mouse line using adiponectin promoter-specific Cre-induced gene inactivation. Control and adipocyte-specific NOS3 knockout (A-NOS3 KO) mice were fed a high-fat diet (HFD). Despite less weight gain, A-NOS3 KO mice exhibited a significant increase in blood pressure after HFD feeding, associated with exacerbated vascular dysfunction and remodelling. A-NOS3 KO mice also showed increased expression of signature markers of inflammation and hypoxia in the PVATs. Among the differentially expressed adipokines, we have observed an upregulation of a novel adipokine, chemerin, in A-NOS3 KO mice. Chemerin was recently reported to link obesity and vascular dysfunction. Treatment with chemerin neutralizing antibody normalized the expression of remodelling markers in the aorta segments cultured in serum from HFD-fed A-NOS3 KO mice ex vivo. CONCLUSION These data suggest that NOS3 in adipocytes is vital in maintaining vascular homeostasis; dysfunction of adipocyte NOS3 contributes to obesity-induced vascular remodelling and hypertension.
Collapse
Affiliation(s)
- Andy W C Man
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Gisela Reifenberg
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Alica Camp
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
19
|
Rashbrook VS, Denti L, Ruhrberg C. Tamoxifen exacerbates morbidity and mortality in male mice receiving medetomidine anaesthesia. Anim Welf 2023; 32:e78. [PMID: 38487465 PMCID: PMC10936365 DOI: 10.1017/awf.2023.98] [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: 04/04/2023] [Revised: 10/20/2023] [Accepted: 11/21/2023] [Indexed: 03/17/2024]
Abstract
Tamoxifen-induced CreER-LoxP recombination is often used to induce spatiotemporally controlled gene deletion in genetically modified mice. Prior work has shown that tamoxifen and tamoxifen-induced CreER activation can have off-target effects that should be controlled. However, it has not yet been reported whether tamoxifen administration, independently of CreER expression, interacts adversely with commonly used anaesthetic drugs such as medetomidine or its enantiomer dexmedetomidine in laboratory mice (Mus musculus). Here, we report a high incidence of urinary plug formation and morbidity in male mice on a mixed C57Bl6/J6 and 129/SvEv background when tamoxifen treatment was followed by ketamine-medetomidine anaesthesia. Medetomidine is therefore contra-indicated for male mice after tamoxifen treatment. As dexmedetomidine causes morbidity and mortality in male mice at higher rates than medetomidine even without tamoxifen treatment, our findings suggest that dexmedetomidine is not a suitable alternative for anaesthesia of male mice after tamoxifen treatment. We conclude that the choice of anaesthetic drug needs to be carefully evaluated in studies using male mice that have undergone tamoxifen treatment for inducing CreER-LoxP recombination.
Collapse
Affiliation(s)
- Victoria S Rashbrook
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
20
|
Lyu QR, Fu K. Tissue-specific Cre driver mice to study vascular diseases. Vascul Pharmacol 2023; 153:107241. [PMID: 37923099 DOI: 10.1016/j.vph.2023.107241] [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: 08/02/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Vascular diseases, including atherosclerosis and abdominal aneurysms, are the primary cause of mortality and morbidity among the elderly worldwide. The life quality of patients is significantly compromised due to inadequate therapeutic approaches and limited drug targets. To expand our comprehension of vascular diseases, gene knockout (KO) mice, especially conditional knockout (cKO) mice, are widely used for investigating gene function and mechanisms of action. The Cre-loxP system is the most common method for generating cKO mice. Numerous Cre driver mice have been established to study the main cell types that compose blood vessels, including endothelial cells, smooth muscle cells, and fibroblasts. Here, we first discuss the characteristics of each layer of the arterial wall. Next, we provide an overview of the representative Cre driver mice utilized for each of the major cell types in the vessel wall and their most recent applications in vascular biology. We then go over Cre toxicity and discuss the practical methods for minimizing Cre interference in experimental outcomes. Finally, we look into the future of tissue-specific Cre drivers by introducing the revolutionary single-cell RNA sequencing and dual recombinase system.
Collapse
Affiliation(s)
- Qing Rex Lyu
- Medical Research Center, Chongqing General Hospital, Chongqing 401147, China; Chongqing Academy of Medical Sciences, Chongqing 401147, China.
| | - Kailong Fu
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| |
Collapse
|
21
|
Ahmed A, Syed JN, Chi L, Wang Y, Perez-Romero C, Lee D, Kocaqi E, Caballero A, Yang J, Escalante-Covarrubias Q, Ishimura A, Suzuki T, Aguilar-Arnal L, Gonzales GB, Kim KH, Delgado-Olguín P. KDM8 epigenetically controls cardiac metabolism to prevent initiation of dilated cardiomyopathy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:174-191. [PMID: 38665902 PMCID: PMC11041705 DOI: 10.1038/s44161-023-00214-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/05/2023] [Indexed: 04/28/2024]
Abstract
Cardiac metabolism is deranged in heart failure, but underlying mechanisms remain unclear. Here, we show that lysine demethylase 8 (Kdm8) maintains an active mitochondrial gene network by repressing Tbx15, thus preventing dilated cardiomyopathy leading to lethal heart failure. Deletion of Kdm8 in mouse cardiomyocytes increased H3K36me2 with activation of Tbx15 and repression of target genes in the NAD+ pathway before dilated cardiomyopathy initiated. NAD+ supplementation prevented dilated cardiomyopathy in Kdm8 mutant mice, and TBX15 overexpression blunted NAD+-activated cardiomyocyte respiration. Furthermore, KDM8 was downregulated in human hearts affected by dilated cardiomyopathy, and higher TBX15 expression defines a subgroup of affected hearts with the strongest downregulation of genes encoding mitochondrial proteins. Thus, KDM8 represses TBX15 to maintain cardiac metabolism. Our results suggest that epigenetic dysregulation of metabolic gene networks initiates myocardium deterioration toward heart failure and could underlie heterogeneity of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Abdalla Ahmed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Jibran Nehal Syed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Lijun Chi
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Yaxu Wang
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Carmina Perez-Romero
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Dorothy Lee
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Physiology, University of Toronto, Toronto, Ontario Canada
| | - Etri Kocaqi
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Amalia Caballero
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Jielin Yang
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Quetzalcoatl Escalante-Covarrubias
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Akihiko Ishimura
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Gerard Bryan Gonzales
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Kyoung-Han Kim
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa and University of Ottawa Heart Institute, Ottawa, Ontario Canada
| | - Paul Delgado-Olguín
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, Ontario Canada
| |
Collapse
|
22
|
O’Brien BJ, Martin KA, Offermanns S. "Cre"ating New Tools for Smooth Muscle Analysis. Arterioscler Thromb Vasc Biol 2023; 43:212-214. [PMID: 36601960 PMCID: PMC10112502 DOI: 10.1161/atvbaha.122.318855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Brendan J. O’Brien
- Departments of Medicine (Cardiovascular Medicine) and Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Kathleen A. Martin
- Departments of Medicine (Cardiovascular Medicine) and Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Bad Nauheim and Center for Molecular Medicine, Goethe University, Frankfurt
| |
Collapse
|
23
|
McCord TJ, Kontos CD. VEGF-C and VE-cadherin: balancing sinusoidal and lymphatic angiogenesis. NATURE CARDIOVASCULAR RESEARCH 2022; 1:976-977. [PMID: 39195911 DOI: 10.1038/s44161-022-00164-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Timothy J McCord
- Cell and Molecular Biology Program, Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
- Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Christopher D Kontos
- Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA.
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|