1
|
Weldrick JJ, Yi R, Megeney LA, Burgon PG. MicroRNA205: A Key Regulator of Cardiomyocyte Transition from Proliferative to Hypertrophic Growth in the Neonatal Heart. Int J Mol Sci 2024; 25:2206. [PMID: 38396885 PMCID: PMC10889831 DOI: 10.3390/ijms25042206] [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] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The mammalian myocardium grows rapidly during early development due to cardiomyocyte proliferation, which later transitions to cell hypertrophy to sustain the heart's postnatal growth. Although this cell transition in the postnatal heart is consistently preserved in mammalian biology, little is known about the regulatory mechanisms that link proliferation suppression with hypertrophy induction. We reasoned that the production of a micro-RNA(s) could serve as a key bridge to permit changes in gene expression that control the changed cell fate of postnatal cardiomyocytes. We used sequential expression analysis to identify miR205 as a micro-RNA that was uniquely expressed at the cessation of cardiomyocyte growth. Cardiomyocyte-specific miR205 deletion animals showed a 35% increase in heart mass by 3 months of age, with commensurate changes in cell cycle and Hippo pathway activity, confirming miR205's potential role in controlling cardiomyocyte proliferation. In contrast, overexpression of miR205 in newborn hearts had little effect on heart size or function, indicating a complex, probably redundant regulatory system. These findings highlight miR205's role in controlling the shift from cardiomyocyte proliferation to hypertrophic development in the postnatal period.
Collapse
Affiliation(s)
- Jonathan J. Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (J.J.W.); (L.A.M.)
| | - Rui Yi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lynn A. Megeney
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (J.J.W.); (L.A.M.)
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, ON K1Y 4E9, Canada
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Patrick G. Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
2
|
Awwad L, Aronheim A. Tumor Progression Reverses Cardiac Hypertrophy and Fibrosis in a Tetracycline-Regulated ATF3 Transgenic Mouse Model. Cells 2023; 12:2289. [PMID: 37759510 PMCID: PMC10528851 DOI: 10.3390/cells12182289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases (CVD) and cancer are the top deadly diseases in the world. Both CVD and cancer have common risk factors; therefore, with the advances in treatment and life span, both diseases may occur simultaneously in patients. It is becoming evident that CVD and cancer are highly connected, establishing a novel discipline known as cardio-oncology. This includes the cardiomyocyte death following any anti-tumor therapy known as cardiotoxicity as well the intricate interplay between heart failure and cancer. Recent studies, using various mouse models, showed that heart failure promotes tumor growth and metastasis spread. Indeed, patients with heart failure were found to be at higher risk of developing malignant diseases. While the effect of heart failure on cancer is well established, little is known regarding the effect of tumors on heart failure. A recent study from our lab has demonstrated that tumor growth and metastasis ameliorate cardiac remodeling in a pressure-overload mouse model. Nevertheless, this study was inconclusive regarding whether tumor growth solely suppresses cardiac remodeling or is able to reverse existing heart failure outcomes as well. Here, we used a regulable transgenic mouse model for cardiac hypertrophy and fibrosis. Cancer cell implantation suppressed cardiac dysfunction and fibrosis as shown using echocardiography, qRT-PCR and fibrosis staining. In addition, tumor growth resulted in an M1 to M2 macrophage switch, which is correlated with cardiac repair. Macrophage depletion using clodronate liposomes completely abrogated the tumors' beneficial effect. This study highly suggests that harnessing tumor paradigms may lead to the development of novel therapeutic strategies for CVDs and fibrosis.
Collapse
Affiliation(s)
| | - Ami Aronheim
- Department of Cell Biology and Cancer Science, Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa P.O. Box 9649, Israel;
| |
Collapse
|
3
|
Nguyen PD, Gooijers I, Campostrini G, Verkerk AO, Honkoop H, Bouwman M, de Bakker DEM, Koopmans T, Vink A, Lamers GEM, Shakked A, Mars J, Mulder AA, Chocron S, Bartscherer K, Tzahor E, Mummery CL, de Boer TP, Bellin M, Bakkers J. Interplay between calcium and sarcomeres directs cardiomyocyte maturation during regeneration. Science 2023; 380:758-764. [PMID: 37200435 DOI: 10.1126/science.abo6718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/20/2023] [Indexed: 05/20/2023]
Abstract
Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.
Collapse
Affiliation(s)
- Phong D Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Iris Gooijers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Experimental Cardiology, University of Amsterdam, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Hessel Honkoop
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Mara Bouwman
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Tim Koopmans
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Gerda E M Lamers
- Core Facility Microscopy, Institute of Biology, Leiden University, Leiden, Netherlands
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jonas Mars
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Sonja Chocron
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Kerstin Bartscherer
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Animal Physiology, Osnabrueck University, Osnabrück, Germany
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Teun P de Boer
- Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
4
|
Shakked A, Petrover Z, Aharonov A, Ghiringhelli M, Umansky KB, Kain D, Elkahal J, Divinsky Y, Nguyen PD, Miyara S, Friedlander G, Savidor A, Zhang L, Perez DE, Sarig R, Lendengolts D, Bueno-Levy H, Kastan N, Levin Y, Bakkers J, Gepstein L, Tzahor E. Redifferentiated cardiomyocytes retain residual dedifferentiation signatures and are protected against ischemic injury. NATURE CARDIOVASCULAR RESEARCH 2023; 2:383-398. [PMID: 37974970 PMCID: PMC10653068 DOI: 10.1038/s44161-023-00250-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 02/09/2023] [Indexed: 11/19/2023]
Abstract
Cardiomyocyte proliferation and dedifferentiation have fueled the field of regenerative cardiology in recent years, whereas the reverse process of redifferentiation remains largely unexplored. Redifferentiation is characterized by the restoration of function lost during dedifferentiation. Previously, we showed that ERBB2-mediated heart regeneration has these two distinct phases: transient dedifferentiation and redifferentiation. Here we survey the temporal transcriptomic and proteomic landscape of dedifferentiation-redifferentiation in adult mouse hearts and reveal that well-characterized dedifferentiation features largely return to normal, although elements of residual dedifferentiation remain, even after the contractile function is restored. These hearts appear rejuvenated and show robust resistance to ischemic injury, even 5 months after redifferentiation initiation. Cardiomyocyte redifferentiation is driven by negative feedback signaling and requires LATS1/2 Hippo pathway activity. Our data reveal the importance of cardiomyocyte redifferentiation in functional restoration during regeneration but also protection against future insult, in what could lead to a potential prophylactic treatment against ischemic heart disease for at-risk patients.
Collapse
Affiliation(s)
- Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Zachary Petrover
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alla Aharonov
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Matteo Ghiringhelli
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Kfir-Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - David Kain
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Elkahal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yalin Divinsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Phong Dang Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Shoval Miyara
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gilgi Friedlander
- Mantoux Bioinformatics Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Lingling Zhang
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Dahlia E. Perez
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rachel Sarig
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daria Lendengolts
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanna Bueno-Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nathaniel Kastan
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, New York, NY, USA
| | - Yishai Levin
- De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lior Gepstein
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
5
|
Lin Q, Kumar S, Kariyawasam U, Yang X, Yang W, Skinner JT, Gao WD, Johns RA. Human Resistin Induces Cardiac Dysfunction in Pulmonary Hypertension. J Am Heart Assoc 2023; 12:e027621. [PMID: 36927008 PMCID: PMC10111547 DOI: 10.1161/jaha.122.027621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 03/18/2023]
Abstract
Background Cardiac failure is the primary cause of death in most patients with pulmonary arterial hypertension (PH). As pleiotropic cytokines, human resistin (Hresistin) and its rodent homolog, resistin-like molecule α, are mechanistically critical to pulmonary vascular remodeling in PH. However, it is still unclear whether activation of these resistin-like molecules can directly cause PH-associated cardiac dysfunction and remodeling. Methods and Results In this study, we detected Hresistin protein in right ventricular (RV) tissue of patients with PH and elevated resistin-like molecule expression in RV tissues of rodents with RV hypertrophy and failure. In a humanized mouse model, cardiac-specific Hresistin overexpression was sufficient to cause cardiac dysfunction and remodeling. Dilated hearts exhibited reduced force development and decreased intracellular Ca2+ transients. In the RV tissues overexpressing Hresistin, the impaired contractility was associated with the suppression of protein kinase A and AMP-activated protein kinase. Mechanistically, Hresistin activation triggered the inflammation mediated by signaling of the key damage-associated molecular pattern molecule high-mobility group box 1, and subsequently induced pro-proliferative Ki67 in RV tissues of the transgenic mice. Intriguingly, an anti-Hresistin human antibody that we generated protected the myocardium from hypertrophy and failure in the rodent PH models. Conclusions Our data indicate that Hresistin is expressed in heart tissues and plays a role in the development of RV dysfunction and maladaptive remodeling through its immunoregulatory activities. Targeting this signaling to modulate cardiac inflammation may offer a promising strategy to treat PH-associated RV hypertrophy and failure in humans.
Collapse
Affiliation(s)
- Qing Lin
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Santosh Kumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Udeshika Kariyawasam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Xiaomei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of AnesthesiologyQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Wei Yang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
- Department of Cardiovascular MedicineXiangya Hospital, Central South UniversityChangshaChina
| | - John T. Skinner
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Wei Dong Gao
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
6
|
Awwad L, Aronheim A. Cardiac Dysfunction Promotes Cancer Progression via Multiple Secreted Factors. Cancer Res 2022; 82:1753-1761. [PMID: 35260887 PMCID: PMC9359722 DOI: 10.1158/0008-5472.can-21-2463] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/14/2021] [Accepted: 03/03/2022] [Indexed: 01/07/2023]
Abstract
Heart failure and cancer are the leading cause of deaths worldwide. While heart failure and cancer have been considered separate diseases, it is becoming evident that they are highly connected and affect each other's outcomes. Recent studies using experimental mouse models have suggested that heart failure promotes tumor progression. The mouse models used involve major irreversible surgery. Here, we induced heart hypertrophy via expression of activating transcription factor 3 (ATF3) in cardiomyocytes, followed by cancer cells' implantation. Tumors developing in ATF3-transgenic mice grew larger and displayed a more highly metastatic phenotype compared with tumors in wild-type mice. To address whether ATF3 expression or the cardiac outcome are necessary for tumor progression, ATF3 expression was turned off after cardiac hypertrophy development followed by cancer cell implantation. The tumor promotion phenotype and the enhancement of metastatic properties were preserved, suggesting that the failing heart per se is sufficient to promote tumor progression. Serum derived from ATF3-transgenic mice enhanced cancer cell proliferation and increased cancer cell metastatic properties in vitro. Using a cytokine array panel, multiple factors responsible for promoting tumor cell proliferation and the metastatic phenotype were identified. Interestingly, the failing heart and the tumor separately and simultaneously contributed to higher levels of these factors in the serum as well as other tissues and organs. These data suggest the existence of intimate cross-talk between the hypertrophied heart and the tumor that is mediated by secreted factors, leading to cancer promotion and disease deterioration. SIGNIFICANCE This work highlights the importance of early diagnosis and treatment of heart failure prior to reaching the irreversible stage that can exacerbate cancer progression.
Collapse
Affiliation(s)
| | - Ami Aronheim
- Corresponding Author: Ami Aronheim, Israel Institute of Technology, 7th Efron St. Bat-Galim, PO Box 9649, Haifa 31096, Israel. Phone: 972-4829-5454; Fax: 972-4829-5225; E-mail:
| |
Collapse
|
7
|
Auchampach J, Han L, Huang GN, Kühn B, Lough JW, O'Meara CC, Payumo AY, Rosenthal NA, Sucov HM, Yutzey KE, Patterson M. Measuring cardiomyocyte cell-cycle activity and proliferation in the age of heart regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H579-H596. [PMID: 35179974 PMCID: PMC8934681 DOI: 10.1152/ajpheart.00666.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
During the past two decades, the field of mammalian myocardial regeneration has grown dramatically, and with this expanded interest comes increasing claims of experimental manipulations that mediate bona fide proliferation of cardiomyocytes. Too often, however, insufficient evidence or improper controls are provided to support claims that cardiomyocytes have definitively proliferated, a process that should be strictly defined as the generation of two de novo functional cardiomyocytes from one original cardiomyocyte. Throughout the literature, one finds inconsistent levels of experimental rigor applied, and frequently the specific data supplied as evidence of cardiomyocyte proliferation simply indicate cell-cycle activation or DNA synthesis, which do not necessarily lead to the generation of new cardiomyocytes. In this review, we highlight potential problems and limitations faced when characterizing cardiomyocyte proliferation in the mammalian heart, and summarize tools and experimental standards, which should be used to support claims of proliferation-based remuscularization. In the end, definitive establishment of de novo cardiomyogenesis can be difficult to prove; therefore, rigorous experimental strategies should be used for such claims.
Collapse
Affiliation(s)
- John Auchampach
- Department of Pharmacology and Toxicology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Lu Han
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Pediatric Cardiology, Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, California
| | - Bernhard Kühn
- Division of Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania
- McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania
| | - John W Lough
- Department of Cell Biology Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Caitlin C O'Meara
- Department of Physiology and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Y Payumo
- Department of Biological Sciences, San José State University, San Jose, California
| | - Nadia A Rosenthal
- The Jackson Laboratory, Bar Harbor, Maine
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
- National Heart and Lung Institute, Imperial College of London, London, United Kingdom
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Katherine E Yutzey
- The Heart Institute, Cincinnati Children's Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Michaela Patterson
- Department of Cell Biology Neurobiology and Anatomy and the Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
8
|
Li Q, Li C, Elnwasany A, Sharma G, An YA, Zhang G, Elhelaly WM, Lin J, Gong Y, Chen G, Wang M, Zhao S, Dai C, Smart CD, Liu J, Luo X, Deng Y, Tan L, Lv SJ, Davidson SM, Locasale JW, Lorenzi PL, Malloy CR, Gillette TG, Vander Heiden MG, Scherer PE, Szweda LI, Fu G, Wang ZV. PKM1 Exerts Critical Roles in Cardiac Remodeling Under Pressure Overload in the Heart. Circulation 2021; 144:712-727. [PMID: 34102853 PMCID: PMC8405569 DOI: 10.1161/circulationaha.121.054885] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metabolic remodeling precedes most alterations during cardiac hypertrophic growth under hemodynamic stress. The elevation of glucose utilization has been recognized as a hallmark of metabolic remodeling. However, its role in cardiac hypertrophic growth and heart failure in response to pressure overload remains to be fully illustrated. Here, we aimed to dissect the role of cardiac PKM1 (pyruvate kinase muscle isozyme 1) in glucose metabolic regulation and cardiac response under pressure overload. METHODS Cardiac-specific deletion of PKM1 was achieved by crossing the floxed PKM1 mouse model with the cardiomyocyte-specific Cre transgenic mouse. PKM1 transgenic mice were generated under the control of tetracycline response elements, and cardiac-specific overexpression of PKM1 was induced by doxycycline administration in adult mice. Pressure overload was triggered by transverse aortic constriction. Primary neonatal rat ventricular myocytes were used to dissect molecular mechanisms. Moreover, metabolomics and nuclear magnetic resonance spectroscopy analyses were conducted to determine cardiac metabolic flux in response to pressure overload. RESULTS We found that PKM1 expression is reduced in failing human and mouse hearts. It is important to note that cardiomyocyte-specific deletion of PKM1 exacerbates cardiac dysfunction and fibrosis in response to pressure overload. Inducible overexpression of PKM1 in cardiomyocytes protects the heart against transverse aortic constriction-induced cardiomyopathy and heart failure. At the mechanistic level, PKM1 is required for the augmentation of glycolytic flux, mitochondrial respiration, and ATP production under pressure overload. Furthermore, deficiency of PKM1 causes a defect in cardiomyocyte growth and a decrease in pyruvate dehydrogenase complex activity at both in vitro and in vivo levels. CONCLUSIONS These findings suggest that PKM1 plays an essential role in maintaining a homeostatic response in the heart under hemodynamic stress.
Collapse
Affiliation(s)
- Qinfeng Li
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chao Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abdallah Elnwasany
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Waleed M. Elhelaly
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Guihao Chen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Meihui Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chongshan Dai
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Charles D. Smart
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Juan Liu
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shuang-Jie Lv
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shawn M. Davidson
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jason W. Locasale
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Craig R. Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luke I. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
9
|
Chronic activation of hexosamine biosynthesis in the heart triggers pathological cardiac remodeling. Nat Commun 2020; 11:1771. [PMID: 32286306 PMCID: PMC7156663 DOI: 10.1038/s41467-020-15640-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/21/2020] [Indexed: 12/21/2022] Open
Abstract
The hexosamine biosynthetic pathway (HBP) plays critical roles in nutrient sensing, stress response, and cell growth. However, its contribution to cardiac hypertrophic growth and heart failure remains incompletely understood. Here, we show that the HBP is induced in cardiomyocytes during hypertrophic growth. Overexpression of Gfat1 (glutamine:fructose-6-phosphate amidotransferase 1), the rate-limiting enzyme of HBP, promotes cardiomyocyte growth. On the other hand, Gfat1 inhibition significantly blunts phenylephrine-induced hypertrophic growth in cultured cardiomyocytes. Moreover, cardiac-specific overexpression of Gfat1 exacerbates pressure overload-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction. Conversely, deletion of Gfat1 in cardiomyocytes attenuates pathological cardiac remodeling in response to pressure overload. Mechanistically, persistent upregulation of the HBP triggers decompensated hypertrophy through activation of mTOR while Gfat1 deficiency shows cardioprotection and a concomitant decrease in mTOR activity. Taken together, our results reveal that chronic upregulation of the HBP under hemodynamic stress induces pathological cardiac hypertrophy and heart failure through persistent activation of mTOR. Metabolic remodeling plays an important role in pathological cardiac hypertrophy. Here, the authors show that hexosamine biosynthetic pathway is elevated in the heart by pressure overload, which contributes to heart failure by persistent activation of mTOR.
Collapse
|
10
|
Honkoop H, de Bakker DE, Aharonov A, Kruse F, Shakked A, Nguyen PD, de Heus C, Garric L, Muraro MJ, Shoffner A, Tessadori F, Peterson JC, Noort W, Bertozzi A, Weidinger G, Posthuma G, Grün D, van der Laarse WJ, Klumperman J, Jaspers RT, Poss KD, van Oudenaarden A, Tzahor E, Bakkers J. Single-cell analysis uncovers that metabolic reprogramming by ErbB2 signaling is essential for cardiomyocyte proliferation in the regenerating heart. eLife 2019; 8:50163. [PMID: 31868166 PMCID: PMC7000220 DOI: 10.7554/elife.50163] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022] Open
Abstract
While the heart regenerates poorly in mammals, efficient heart regeneration occurs in zebrafish. Studies in zebrafish have resulted in a model in which preexisting cardiomyocytes dedifferentiate and reinitiate proliferation to replace the lost myocardium. To identify which processes occur in proliferating cardiomyocytes we have used a single-cell RNA-sequencing approach. We uncovered that proliferating border zone cardiomyocytes have very distinct transcriptomes compared to the nonproliferating remote cardiomyocytes and that they resemble embryonic cardiomyocytes. Moreover, these cells have reduced expression of mitochondrial genes and reduced mitochondrial activity, while glycolysis gene expression and glucose uptake are increased, indicative for metabolic reprogramming. Furthermore, we find that the metabolic reprogramming of border zone cardiomyocytes is induced by Nrg1/ErbB2 signaling and is important for their proliferation. This mechanism is conserved in murine hearts in which cardiomyocyte proliferation is induced by activating ErbB2 signaling. Together these results demonstrate that glycolysis regulates cardiomyocyte proliferation during heart regeneration. Heart attacks are a common cause of death in the Western world. During a heart attack, oxygen levels in the affected part of the heart decrease, which causes heart muscle cells to die. In humans the dead cells are replaced by a permanent scar that stabilizes the injury but does not completely heal it. As a result, individuals have a lower quality of life after a heart attack and are more likely to die from a subsequent attack. Unlike humans, zebrafish are able to regenerate their hearts after injury: heart muscle cells close to a wound divide to produce new cells that slowly replace the scar tissue and restore normal function to the area. It remains unclear, however, what stimulates the heart muscle cells of zebrafish to start dividing. To address this question, Honkoop, de Bakker et al. used a technique called single-cell sequencing to study heart muscle cells in wounded zebrafish hearts. The experiments identified a group of heart muscle cells close to the site of the wound that multiplied to repair the damage. This group of cells had altered their metabolism compared to other heart muscle cells so that they relied on a pathway called glycolysis to produce the energy and building blocks they needed to proliferate. Blocking glycolysis impaired the ability of the heart muscle cells to divide, indicating that this switch is necessary for the heart to regenerate. Further experiments showed that a signaling cascade, which includes the molecules Nrg1 and ErbB2, induces heart muscle cells in both zebrafish and mouse hearts to switch to glycolysis and undergo division. These findings indicate that activating glycolysis in heart muscle cells may help to stimulate the heart to regenerate after a heart attack or other injury. The next step following on from this work is to develop methods to activate glycolysis and promote cell division in injured hearts.
Collapse
Affiliation(s)
- Hessel Honkoop
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Dennis Em de Bakker
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Alla Aharonov
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Fabian Kruse
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Phong D Nguyen
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Cecilia de Heus
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Laurence Garric
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Mauro J Muraro
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Adam Shoffner
- Regeneration Next, Department of Cell Biology, Duke University Medical Center, Durham, United States
| | - Federico Tessadori
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Joshua Craiger Peterson
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wendy Noort
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Alberto Bertozzi
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - George Posthuma
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Dominic Grün
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Willem J van der Laarse
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Kenneth D Poss
- Regeneration Next, Department of Cell Biology, Duke University Medical Center, Durham, United States
| | | | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
11
|
Frank DU, Sutcliffe MD, Saucerman JJ. Network-based predictions of in vivo cardiac hypertrophy. J Mol Cell Cardiol 2018; 121:180-189. [PMID: 30030017 DOI: 10.1016/j.yjmcc.2018.07.243] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
Cardiac hypertrophy is a common response of cardiac myocytes to stress and a predictor of heart failure. While in vitro cell culture studies have identified numerous molecular mechanisms driving hypertrophy, it is unclear to what extent these mechanisms can be integrated into a consistent framework predictive of in vivo phenotypes. To address this question, we investigate the degree to which an in vitro-based, manually curated computational model of the hypertrophy signaling network is able to predict in vivo hypertrophy of 52 cardiac-specific transgenic mice. After minor revisions motivated by in vivo literature, the model concordantly predicts the qualitative responses of 78% of output species and 69% of signaling intermediates within the network model. Analysis of four double-transgenic mouse models reveals that the computational model robustly predicts hypertrophic responses in mice subjected to multiple, simultaneous perturbations. Thus the model provides a framework with which to mechanistically integrate data from multiple laboratories and experimental systems to predict molecular regulation of cardiac hypertrophy.
Collapse
Affiliation(s)
- Deborah U Frank
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States; Department of Pediatrics, University of Virginia, HSC Box 800386, Charlottesville 22908-0386, VA, United States.
| | - Matthew D Sutcliffe
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States; Department of Pediatrics, University of Virginia, HSC Box 800386, Charlottesville 22908-0386, VA, United States.
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States.
| |
Collapse
|
12
|
Cardiac-specific inducible overexpression of human plasma membrane Ca 2+ ATPase 4b is cardioprotective and improves survival in mice following ischemic injury. Clin Sci (Lond) 2018; 132:641-654. [PMID: 29487197 DOI: 10.1042/cs20171337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/09/2023]
Abstract
Background: Heart failure (HF) is associated with reduced expression of plasma membrane Ca2+-ATPase 4 (PMCA4). Cardiac-specific overexpression of human PMCA4b in mice inhibited nNOS activity and reduced cardiac hypertrophy by inhibiting calcineurin. Here we examine temporally regulated cardiac-specific overexpression of hPMCA4b in mouse models of myocardial ischemia reperfusion injury (IRI) ex vivo, and HF following experimental myocardial infarction (MI) in vivoMethods and results: Doxycycline-regulated cardiomyocyte-specific overexpression and activity of hPMCA4b produced adaptive changes in expression levels of Ca2+-regulatory genes, and induced hypertrophy without significant differences in Ca2+ transients or diastolic Ca2+ concentrations. Total cardiac NOS and nNOS-specific activities were reduced in mice with cardiac overexpression of hPMCA4b while nNOS, eNOS and iNOS protein levels did not differ. hMPCA4b-overexpressing mice also exhibited elevated systolic blood pressure vs. controls, with increased contractility and lusitropy in vivo In isolated hearts undergoing IRI, hPMCA4b overexpression was cardioprotective. NO donor-treated hearts overexpressing hPMCA4b showed reduced LVDP and larger infarct size versus vehicle-treated hearts undergoing IRI, demonstrating that the cardioprotective benefits of hPMCA4b-repressed nNOS are lost by restoring NO availability. Finally, both pre-existing and post-MI induction of hPMCA4b overexpression reduced infarct expansion and improved survival from HF.Conclusions: Cardiac PMCA4b regulates nNOS activity, cardiac mass and contractility, such that PMCA4b overexpression preserves cardiac function following IRI, heightens cardiac performance and limits infarct progression, cardiac hypertrophy and HF, even when induced late post-MI. These data identify PMCA4b as a novel therapeutic target for IRI and HF.
Collapse
|
13
|
He Z, Grunewald M, Dor Y, Keshet E. VEGF regulates relative allocation of Isl1 + cardiac progenitors to myocardial and endocardial lineages. Mech Dev 2016; 142:40-49. [PMID: 27794491 DOI: 10.1016/j.mod.2016.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 01/07/2023]
Abstract
A fundamental issue in organogenesis is how dichotomous fate decisions are made securing proper allocation of multipotent progenitors to their respective descendants. Previous lineage tracing analyses showing Isl1+/VEGFR2+ cardiac progenitors in the second heart field give rise to both endocardium and myocardium suggest VEGF plays a role in this fate decision, conceivably promoting an endocardial fate. Isl1+ multipotent progenitors and lineage-committed descendants thereof were visualized and quantified within their transition zone in the outflow tract. Forced VEGF expression during the critical E8.5-E10.5 interval tilted the balance between myocardial- and endocardial-committed progenitors towards the latter, culminating in generation of surplus endocardium developing at the expense of a much thinner myocardium. Experiments ruled-out that surplus endocardium is due to VEGF-induced endocardial proliferation and that reduced myocardium is due to myocardial apoptosis. Inducing VEGF after most Isl1+ progenitors have been exhausted had no effect on the normal endocardia/myocardial ratio but instead produced an unrelated coronary phenotype. Together, these results uncover a novel role for VEGF in controlling proper allocation of Isl1+ cardiac progenitors to their respective descending lineages.
Collapse
Affiliation(s)
- Zhiheng He
- Department of Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Myriam Grunewald
- Department of Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yuval Dor
- Department of Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Eli Keshet
- Department of Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
14
|
Mayrhofer M, Mione M. The Toolbox for Conditional Zebrafish Cancer Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:21-59. [PMID: 27165348 DOI: 10.1007/978-3-319-30654-4_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here we describe the conditional zebrafish cancer toolbox, which allows for fine control of the expression of oncogenes or downregulation of tumor suppressors at the spatial and temporal level. Methods such as the Gal4/UAS or the Cre/lox systems paved the way to the development of elegant tumor models, which are now being used to study cancer cell biology, clonal evolution, identification of cancer stem cells and anti-cancer drug screening. Combination of these tools, as well as novel developments such as the promising genome editing system through CRISPR/Cas9 and clever application of light reactive proteins will enable the development of even more sophisticated zebrafish cancer models. Here, we introduce this growing toolbox of conditional transgenic approaches, discuss its current application in zebrafish cancer models and provide an outlook on future perspectives.
Collapse
Affiliation(s)
- Marie Mayrhofer
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Marina Mione
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
15
|
Kraut B, Maier HJ, Kókai E, Fiedler K, Boettger T, Illing A, Kostin S, Walther P, Braun T, Wirth T. Cardiac-Specific Activation of IKK2 Leads to Defects in Heart Development and Embryonic Lethality. PLoS One 2015; 10:e0141591. [PMID: 26539991 PMCID: PMC4634958 DOI: 10.1371/journal.pone.0141591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/09/2015] [Indexed: 11/24/2022] Open
Abstract
The transcription factor NF-κB has been associated with a range of pathological conditions of the heart, mainly based on its function as a master regulator of inflammation and pro-survival factor. Here, we addressed the question what effects activation of NF-κB can have during murine heart development. We expressed a constitutively active (CA) mutant of IKK2, the kinase activating canonical NF-κB signaling, specifically in cardiomyocytes under the control of the α-myosin heavy chain promoter. Expression of IKK2-CA resulted in embryonic lethality around E13. Embryos showed defects in compact zone formation and the contractile apparatus, and overall were characterized by widespread inflammation with infiltration of myeloid cells. Gene expression analysis suggested an interferon type I signature, with increased expression of interferon regulatory factors. While apoptosis of cardiomyocytes was only increased at later stages, their proliferation was decreased early on, providing an explanation for the disturbed compact zone formation. Mechanistically, this could be explained by activation of the JAK/STAT axis and increased expression of the cell cycle inhibitor p21. A rescue experiment with an IκBα superrepressor demonstrated that the phenotype was dependent on NF-κB. We conclude that activation of NF-κB is detrimental during normal heart development due to excessive activation of pro-inflammatory pathways.
Collapse
Affiliation(s)
- Bärbel Kraut
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Harald J. Maier
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Enikö Kókai
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Katja Fiedler
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Thomas Boettger
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Annett Illing
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Sawa Kostin
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Paul Walther
- Core Facility Electron Microscopy, University of Ulm, Ulm, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
- * E-mail:
| |
Collapse
|
16
|
Cannon L, Yu ZY, Marciniec T, Waardenberg AJ, Iismaa SE, Nikolova-Krstevski V, Neist E, Ohanian M, Qiu MR, Rainer S, Harvey RP, Feneley MP, Graham RM, Fatkin D. Irreversible triggers for hypertrophic cardiomyopathy are established in the early postnatal period. J Am Coll Cardiol 2015; 65:560-9. [PMID: 25677315 DOI: 10.1016/j.jacc.2014.10.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is caused by mutations in sarcomere protein genes, and left ventricular hypertrophy (LVH) develops as an adaptive response to sarcomere dysfunction. It remains unclear whether persistent expression of the mutant gene is required for LVH or whether early gene expression acts as an immutable inductive trigger. OBJECTIVES The aim of this study was to use a regulatable murine model of HCM to study the reversibility of pathological LVH. METHODS The authors generated a double-transgenic mouse model, tTAxαMHCR403Q, in which expression of the HCM-causing Arg403Gln mutation in the α-myosin heavy chain (MHC) gene is inhibited by doxycycline administration. Cardiac structure and function were evaluated in groups of mice that received doxycycline for varying periods from 0 to 40 weeks of age. RESULTS Untreated tTAxαMHCR403Q mice showed increased left ventricular (LV) mass, contractile dysfunction, myofibrillar disarray, and fibrosis. In contrast, mice treated with doxycycline from conception to 6 weeks had markedly less LVH and fibrosis at 40 weeks. Transgene inhibition from 6 weeks reduced fibrosis but did not prevent LVH or functional changes. There were no differences in LV parameters at 40 weeks between mice with transgene inhibition from 20 weeks and mice with continuous transgene expression. CONCLUSIONS These findings highlight the critical role of the early postnatal period in HCM pathogenesis and suggest that mutant sarcomeres manifest irreversible cardiomyocyte defects that induce LVH. In HCM, mutation-silencing therapies are likely to be ineffective for hypertrophy regression and would have to be administered very early in life to prevent hypertrophy development.
Collapse
Affiliation(s)
- Leah Cannon
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Ze-Yan Yu
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Tadeusz Marciniec
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Ashley J Waardenberg
- Cardiac Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia
| | - Vesna Nikolova-Krstevski
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia
| | - Elysia Neist
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Monique Ohanian
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Min Ru Qiu
- Anatomical Pathology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Stephen Rainer
- Anatomical Pathology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Richard P Harvey
- Cardiac Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Australia
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia.
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia; Faculty of Medicine, University of New South Wales, Kensington, Australia; Cardiology Department, St. Vincent's Hospital, Darlinghurst, Australia.
| |
Collapse
|
17
|
ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol 2015; 17:627-38. [PMID: 25848746 DOI: 10.1038/ncb3149] [Citation(s) in RCA: 502] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 03/05/2015] [Indexed: 12/14/2022]
Abstract
The murine neonatal heart can regenerate after injury through cardiomyocyte (CM) proliferation, although this capacity markedly diminishes after the first week of life. Neuregulin-1 (NRG1) administration has been proposed as a strategy to promote cardiac regeneration. Here, using loss- and gain-of-function genetic tools, we explore the role of the NRG1 co-receptor ERBB2 in cardiac regeneration. NRG1-induced CM proliferation diminished one week after birth owing to a reduction in ERBB2 expression. CM-specific Erbb2 knockout revealed that ERBB2 is required for CM proliferation at embryonic/neonatal stages. Induction of a constitutively active ERBB2 (caERBB2) in neonatal, juvenile and adult CMs resulted in cardiomegaly, characterized by extensive CM hypertrophy, dedifferentiation and proliferation, differentially mediated by ERK, AKT and GSK3β/β-catenin signalling pathways. Transient induction of caERBB2 following myocardial infarction triggered CM dedifferentiation and proliferation followed by redifferentiation and regeneration. Thus, ERBB2 is both necessary for CM proliferation and sufficient to reactivate postnatal CM proliferative and regenerative potentials.
Collapse
|
18
|
Cardiac arrhythmia induced by genetic silencing of 'funny' (f) channels is rescued by GIRK4 inactivation. Nat Commun 2014; 5:4664. [PMID: 25144323 PMCID: PMC4207211 DOI: 10.1038/ncomms5664] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/14/2014] [Indexed: 01/01/2023] Open
Abstract
The mechanisms underlying cardiac automaticity are still incompletely
understood and controversial. Here we report the complete conditional and
time-controlled silencing of the "funny" current
(If) by expression of a dominant-negative,
non-conductive HCN4-channel subunit (hHCN4-AYA). Heart-specific
If silencing caused altered
[Ca2+]i release and Ca2+ handling in the
sinoatrial node, impaired pacemaker activity, and symptoms reminiscent of severe
human disease of pacemaking. The effects of If
silencing critically depended on the activity of the autonomic nervous system.
We were able to rescue the failure of impulse generation and conduction by
additional genetic deletion of cardiac muscarinic G-protein-activated (GIRK4)
channels in If-deficient mice without impairing
heartbeat regulation. Our study establishes the role of f-channels in cardiac
automaticity and indicates that arrhythmia related to HCN
loss-of-function may be managed by pharmacological or genetic inhibition of
GIRK4 channels, thus offering a new therapeutic strategy for the treatment of
heart rhythm diseases.
Collapse
|
19
|
Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, Criollo A, Luo X, Tan W, Jiang N, Lehrman MA, Rothermel BA, Lee AH, Lavandero S, Mammen PPA, Ferdous A, Gillette TG, Scherer PE, Hill JA. Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell 2014; 156:1179-1192. [PMID: 24630721 DOI: 10.1016/j.cell.2014.01.014] [Citation(s) in RCA: 328] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 11/22/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022]
Abstract
The hexosamine biosynthetic pathway (HBP) generates uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) for glycan synthesis and O-linked GlcNAc (O-GlcNAc) protein modifications. Despite the established role of the HBP in metabolism and multiple diseases, regulation of the HBP remains largely undefined. Here, we show that spliced X-box binding protein 1 (Xbp1s), the most conserved signal transducer of the unfolded protein response (UPR), is a direct transcriptional activator of the HBP. We demonstrate that the UPR triggers HBP activation via Xbp1s-dependent transcription of genes coding for key, rate-limiting enzymes. We further establish that this previously unrecognized UPR-HBP axis is triggered in a variety of stress conditions. Finally, we demonstrate a physiologic role for the UPR-HBP axis by showing that acute stimulation of Xbp1s in heart by ischemia/reperfusion confers robust cardioprotection in part through induction of the HBP. Collectively, these studies reveal that Xbp1s couples the UPR to the HBP to protect cells under stress.
Collapse
Affiliation(s)
- Zhao V Wang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ningguo Gao
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zully Pedrozo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Advanced Center for Chronic Diseases (ACCDiS) and Centro Estudios Moleculares de la Celula, Facultad Ciencias Quimicas y Farmaceuticas and Facultad Medicina, Universidad de Chile, Santiago, Chile; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Dan L Li
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cyndi R Morales
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alfredo Criollo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Advanced Center for Chronic Diseases (ACCDiS) and Centro Estudios Moleculares de la Celula, Facultad Ciencias Quimicas y Farmaceuticas and Facultad Medicina, Universidad de Chile, Santiago, Chile; Dental Science Research Institute, Facultad de Odontologia, Universidad de Chile, Santiago, Chile
| | - Xiang Luo
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Tan
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nan Jiang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mark A Lehrman
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ann-Hwee Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Sergio Lavandero
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Advanced Center for Chronic Diseases (ACCDiS) and Centro Estudios Moleculares de la Celula, Facultad Ciencias Quimicas y Farmaceuticas and Facultad Medicina, Universidad de Chile, Santiago, Chile; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Pradeep P A Mammen
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anwarul Ferdous
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas G Gillette
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
20
|
Gordon O, He Z, Gilon D, Gruener S, Pietranico-Cole S, Oppenheim A, Keshet E. A transgenic platform for testing drugs intended for reversal of cardiac remodeling identifies a novel 11βHSD1 inhibitor rescuing hypertrophy independently of re-vascularization. PLoS One 2014; 9:e92869. [PMID: 24667808 PMCID: PMC3965501 DOI: 10.1371/journal.pone.0092869] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/27/2014] [Indexed: 12/19/2022] Open
Abstract
Rationale Rescuing adverse myocardial remodeling is an unmet clinical goal and, correspondingly, pharmacological means for its intended reversal are urgently needed. Objectives To harness a newly-developed experimental model recapitulating progressive heart failure development for the discovery of new drugs capable of reversing adverse remodeling. Methods and Results A VEGF-based conditional transgenic system was employed in which an induced perfusion deficit and a resultant compromised cardiac function lead to progressive remodeling and eventually heart failure. Ability of candidate drugs administered at sequential remodeling stages to reverse hypertrophy, enlarged LV size and improve cardiac function was monitored. Arguing for clinical relevance of the experimental system, clinically-used drugs operating on the Renin-Angiotensin-Aldosterone-System (RAAS), namely, the ACE inhibitor Enalapril and the direct renin inhibitor Aliskerin fully reversed remodeling. Remodeling reversal by these drugs was not accompanied by neovascularization and reached a point-of-no-return. Similarly, the PPARγ agonist Pioglitazone was proven capable of reversing all aspects of cardiac remodeling without affecting the vasculature. Extending the arsenal of remodeling-reversing drugs to pathways other than RAAS, a specific inhibitor of 11β-hydroxy-steroid dehydrogenase type 1 (11β HSD1), a key enzyme required for generating active glucocorticoids, fully rescued myocardial hypertrophy. This was associated with mitigating the hypertrophy-associated gene signature, including reversing the myosin heavy chain isoform switch but in a pattern distinguishable from that associated with neovascularization-induced reversal. Conclusions A system was developed suitable for identifying novel remodeling-reversing drugs operating in different pathways and for gaining insights into their mechanisms of action, exemplified here by uncoupling their vascular affects.
Collapse
Affiliation(s)
- Oren Gordon
- Departments of Developmental Biology and Cancer Research, The Hebrew University–Hadassah University Hospital, Jerusalem, Israel
| | - Zhiheng He
- Departments of Developmental Biology and Cancer Research, The Hebrew University–Hadassah University Hospital, Jerusalem, Israel
| | - Dan Gilon
- Department of Cardiology, The Hebrew University–Hadassah University Hospital, Jerusalem, Israel
| | - Sabine Gruener
- Department of Metabolic and Vascular Disease, Hoffmann-La Roche Pharmaceuticals, Basel, Switzerland
| | - Sherrie Pietranico-Cole
- Department of Metabolic and Vascular Disease, Hoffmann-La Roche Pharmaceuticals, Basel, Switzerland
| | - Amit Oppenheim
- Department of Cardiology, The Hebrew University–Hadassah University Hospital, Jerusalem, Israel
| | - Eli Keshet
- Departments of Developmental Biology and Cancer Research, The Hebrew University–Hadassah University Hospital, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
21
|
Dalal S, Zha Q, Daniels CR, Steagall RJ, Joyner WL, Gadeau AP, Singh M, Singh K. Osteopontin stimulates apoptosis in adult cardiac myocytes via the involvement of CD44 receptors, mitochondrial death pathway, and endoplasmic reticulum stress. Am J Physiol Heart Circ Physiol 2014; 306:H1182-91. [PMID: 24531809 DOI: 10.1152/ajpheart.00954.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Increased osteopontin (OPN) expression associates with increased myocyte apoptosis and myocardial dysfunction. The objective of this study was to identify the receptor for OPN and get insight into the mechanism by which OPN induces cardiac myocyte apoptosis. Adult rat ventricular myocytes (ARVMs) and transgenic mice expressing OPN in a myocyte-specific manner were used for in vitro and in vivo studies. Treatment with purified OPN (20 nM) protein or adenoviral-mediated OPN expression induced apoptosis in ARVMs. OPN co-immunoprecipitated with CD44 receptors, not with β1 or β3 integrins. Proximity ligation assay confirmed interaction of OPN with CD44 receptors. Neutralizing anti-CD44 antibodies inhibited OPN-stimulated apoptosis. OPN activated JNKs and increased expression of Bax and levels of cytosolic cytochrome c, suggesting involvement of mitochondrial death pathway. OPN increased endoplasmic reticulum (ER) stress, as evidenced by increased expression of Gadd153 and activation of caspase-12. Inhibition of JNKs using SP600125 or ER stress using salubrinal or caspase-12 inhibitor significantly reduced OPN-stimulated apoptosis. Expression of OPN in adult mouse heart in myocyte-specific manner associated with decreased left ventricular function and increased myocyte apoptosis. In the heart, OPN expression increased JNKs and caspase-12 activities, and expression of Bax and Gadd153. Thus, OPN, acting via CD44 receptors, induces apoptosis in myocytes via the involvement of mitochondrial death pathway and ER stress.
Collapse
Affiliation(s)
- Suman Dalal
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Montano MM, Desjardins CL, Doughman YQ, Hsieh YH, Hu Y, Bensinger HM, Wang C, Stelzer JE, Dick TE, Hoit BD, Chandler MP, Yu X, Watanabe M. Inducible re-expression of HEXIM1 causes physiological cardiac hypertrophy in the adult mouse. Cardiovasc Res 2013; 99:74-82. [PMID: 23585471 PMCID: PMC3687752 DOI: 10.1093/cvr/cvt086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/28/2013] [Accepted: 04/01/2013] [Indexed: 01/06/2023] Open
Abstract
AIMS The transcription factor hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) regulates myocardial vascularization and growth during cardiogenesis. Our aim was to determine whether HEXIM1 also has a beneficial role in modulating vascularization, myocardial growth, and function within the adult heart. METHODS AND RESULTS To achieve our objective, we created and investigated a mouse line wherein HEXIM1 was re-expressed in adult cardiomyocytes to levels found in the foetal heart. Our findings support a beneficial role for HEXIM1 through increased vascularization, myocardial growth, and increased ejection fraction within the adult heart. HEXIM1 re-expression induces angiogenesis, that is, essential for physiological hypertrophy and maintenance of cardiac function. The ability of HEXIM1 to co-ordinate processes associated with physiological hypertrophy may be attributed to HEXIM1 regulation of other transcription factors (HIF-1-α, c-Myc, GATA4, and PPAR-α) that, in turn, control many genes involved in myocardial vascularization, growth, and metabolism. Moreover, the mechanism for HEXIM1-induced physiological hypertrophy appears to be distinct from that involving the PI3K/AKT pathway. CONCLUSION HEXIM1 re-expression results in the induction of angiogenesis that allows for the co-ordination of tissue growth and angiogenesis during physiological hypertrophy.
Collapse
Affiliation(s)
- Monica M. Montano
- Department of Pharmacology, Case Western Reserve University School of Medicine, H.G. Wood Bldg. W307, 2109 Adelbert Road, Cleveland, OH 44106, USA
| | - Candida L. Desjardins
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland, OH 44106, USA
| | - Yong Qui Doughman
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Genetics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Anatomy, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Yee-Hsee Hsieh
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Yanduan Hu
- Department of Pharmacology, Case Western Reserve University School of Medicine, H.G. Wood Bldg. W307, 2109 Adelbert Road, Cleveland, OH 44106, USA
| | - Heather M. Bensinger
- Department of Pharmacology, Case Western Reserve University School of Medicine, H.G. Wood Bldg. W307, 2109 Adelbert Road, Cleveland, OH 44106, USA
| | - Connie Wang
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Genetics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Anatomy, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Julian E. Stelzer
- Department of Physiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Thomas E. Dick
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian D. Hoit
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Margaret P. Chandler
- Department of Physiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Yu
- Department of Biomedical Engineering, Case Western Reserve University School of Engineering, Cleveland, OH 44106, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Genetics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Anatomy, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
23
|
Ziegler T, Horstkotte J, Schwab C, Pfetsch V, Weinmann K, Dietzel S, Rohwedder I, Hinkel R, Gross L, Lee S, Hu J, Soehnlein O, Franz WM, Sperandio M, Pohl U, Thomas M, Weber C, Augustin HG, Fässler R, Deutsch U, Kupatt C. Angiopoietin 2 mediates microvascular and hemodynamic alterations in sepsis. J Clin Invest 2013; 123:66549. [PMID: 23863629 PMCID: PMC3726157 DOI: 10.1172/jci66549] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 05/06/2013] [Indexed: 01/22/2023] Open
Abstract
Septic shock is characterized by increased vascular permeability and hypotension despite increased cardiac output. Numerous vasoactive cytokines are upregulated during sepsis, including angiopoietin 2 (ANG2), which increases vascular permeability. Here we report that mice engineered to inducibly overexpress ANG2 in the endothelium developed sepsis-like hemodynamic alterations, including systemic hypotension, increased cardiac output, and dilatory cardiomyopathy. Conversely, mice with cardiomyocyte-restricted ANG2 overexpression failed to develop hemodynamic alterations. Interestingly, the hemodynamic alterations associated with endothelial-specific overexpression of ANG2 and the loss of capillary-associated pericytes were reversed by intravenous injections of adeno-associated viruses (AAVs) transducing cDNA for angiopoietin 1, a TIE2 ligand that antagonizes ANG2, or AAVs encoding PDGFB, a chemoattractant for pericytes. To confirm the role of ANG2 in sepsis, we i.p. injected LPS into C57BL/6J mice, which rapidly developed hypotension, acute pericyte loss, and increased vascular permeability. Importantly, ANG2 antibody treatment attenuated LPS-induced hemodynamic alterations and reduced the mortality rate at 36 hours from 95% to 61%. These data indicate that ANG2-mediated microvascular disintegration contributes to septic shock and that inhibition of the ANG2/TIE2 interaction during sepsis is a potential therapeutic target.
Collapse
Affiliation(s)
- Tilman Ziegler
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Jan Horstkotte
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Claudia Schwab
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Vanessa Pfetsch
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Karolina Weinmann
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Steffen Dietzel
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Ina Rohwedder
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Rabea Hinkel
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Lisa Gross
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Seungmin Lee
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Junhao Hu
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Oliver Soehnlein
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Wolfgang M. Franz
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Markus Sperandio
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Ulrich Pohl
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Markus Thomas
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Christian Weber
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Hellmut G. Augustin
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Reinhard Fässler
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Urban Deutsch
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| | - Christian Kupatt
- Medizinische Klinik und Poliklinik I, Klinikum Großhadern, Ludwig Maximilians University, Munich, Germany.
Theodor Kocher Institute, Bern, Switzerland.
Walter Brendel Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany.
Department for Molecular Medicine, Max-Planck Institute for Biochemistry, Martinsried, Germany.
DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
Joint Research Division Vascular Biology, Medical Faculty Mannheim (Centre for Biomedicine and Medical Technology Mannheim) and German Cancer Research Center (DKFZ–Zentrum für Molekulare Biologie der Universität Heidelberg Alliance), Heidelberg, Germany.
Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany.
Discovery Oncology, Pharma Research and Early Development, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
24
|
Cardiomyocyte-specific IκB kinase (IKK)/NF-κB activation induces reversible inflammatory cardiomyopathy and heart failure. Proc Natl Acad Sci U S A 2012; 109:11794-9. [PMID: 22753500 DOI: 10.1073/pnas.1116584109] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Inflammation is a major factor in heart disease. IκB kinase (IKK) and its downstream target NF-κB are regulators of inflammation and are activated in cardiac disorders, but their precise contributions and targets are unclear. We analyzed IKK/NF-κB function in the heart by a gain-of-function approach, generating an inducible transgenic mouse model with cardiomyocyte-specific expression of constitutively active IKK2. In adult animals, IKK2 activation led to inflammatory dilated cardiomyopathy and heart failure. Transgenic hearts showed infiltration with CD11b(+) cells, fibrosis, fetal reprogramming, and atrophy of myocytes with strong constitutively active IKK2 expression. Upon transgene inactivation, the disease was reversible even at an advanced stage. IKK-induced cardiomyopathy was dependent on NF-κB activation, as in vivo expression of IκBα superrepressor, an inhibitor of NF-κB, prevented the development of disease. Gene expression and proteomic analyses revealed enhanced expression of inflammatory cytokines, and an IFN type I signature with activation of the IFN-stimulated gene 15 (ISG15) pathway. In that respect, IKK-induced cardiomyopathy resembled Coxsackievirus-induced myocarditis, during which the NF-κB and ISG15 pathways were also activated. Vice versa, in cardiomyocytes lacking the regulatory subunit of IKK (IKKγ/NEMO), the induction of ISG15 was attenuated. We conclude that IKK/NF-κB activation in cardiomyocytes is sufficient to cause cardiomyopathy and heart failure by inducing an excessive inflammatory response and myocyte atrophy.
Collapse
|
25
|
Vinet L, Pezet M, Bito V, Briec F, Biesmans L, Rouet-Benzineb P, Gellen B, Prévilon M, Chimenti S, Vilaine JP, Charpentier F, Sipido KR, Mercadier JJ. Cardiac FKBP12.6 overexpression protects against triggered ventricular tachycardia in pressure overloaded mouse hearts. Basic Res Cardiol 2012; 107:246. [DOI: 10.1007/s00395-012-0246-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/23/2011] [Accepted: 01/19/2012] [Indexed: 10/14/2022]
|
26
|
Elsherif L, Wang X, Grachoff M, Wolska BM, Geenen DL, O'Bryan JP. Cardiac-specific expression of the tetracycline transactivator confers increased heart function and survival following ischemia reperfusion injury. PLoS One 2012; 7:e30129. [PMID: 22272284 PMCID: PMC3260203 DOI: 10.1371/journal.pone.0030129] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 12/13/2011] [Indexed: 11/18/2022] Open
Abstract
Mice expressing the tetracycline transactivator (tTA) transcription factor driven by the rat α-myosin heavy chain promoter (α-MHC-tTA) are widely used to dissect the molecular mechanisms involved in cardiac development and disease. However, these α-MHC-tTA mice exhibit a gain-of-function phenotype consisting of robust protection against ischemia/reperfusion injury in both in vitro and in vivo models in the absence of associated cardiac hypertrophy or remodeling. Cardiac function, as assessed by echocardiography, did not differ between α-MHC-tTA and control animals, and there were no noticeable differences observed between the two groups in HW/TL ratio or LV end-diastolic and end-systolic dimensions. Protection against ischemia/reperfusion injury was assessed using isolated perfused hearts where α-MHC-tTA mice had robust protection against ischemia/reperfusion injury which was not blocked by pharmacological inhibition of PI3Ks with LY294002. Furthermore, α-MHC-tTA mice subjected to coronary artery ligation exhibited significantly reduced infarct size compared to control animals. Our findings reveal that α-MHC-tTA transgenic mice exhibit a gain-of-function phenotype consisting of robust protection against ischemia/reperfusion injury similar to cardiac pre- and post-conditioning effects. However, in contrast to classical pre- and post-conditioning, the α-MHC-tTA phenotype is not inhibited by the classic preconditioning inhibitor LY294002 suggesting involvement of a non-PI3K-AKT signaling pathway in this phenotype. Thus, further study of the α-MHC-tTA model may reveal novel molecular targets for therapeutic intervention during ischemic injury.
Collapse
Affiliation(s)
- Laila Elsherif
- Department of Pharmacology, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
| | - Xuerong Wang
- Department of Pharmacology, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
| | - Milana Grachoff
- Section of Cardiology, Department of Medicine, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
| | - Beata M. Wolska
- Section of Cardiology, Department of Medicine, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- Department of Physiology and Biophysics, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- Center for Cardiovascular Research, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
| | - David L. Geenen
- Section of Cardiology, Department of Medicine, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- Department of Physiology and Biophysics, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- Center for Cardiovascular Research, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
| | - John P. O'Bryan
- Department of Pharmacology, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- Center for Cardiovascular Research, College of Medicine, University of Illinois-Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
27
|
Lai NC, Tang T, Gao MH, Saito M, Miyanohara A, Hammond HK. Improved function of the failing rat heart by regulated expression of insulin-like growth factor I via intramuscular gene transfer. Hum Gene Ther 2012; 23:255-61. [PMID: 22017392 DOI: 10.1089/hum.2011.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Current methods of gene transfer for heart disease include injection into heart muscle or intracoronary coronary delivery, approaches that typically provide limited expression and are cumbersome to apply. To circumvent these problems, we selected a transgene, insulin-like growth factor-I (IGF-I), which may, in theory, have favorable effects on heart function when secreted from a remote site. We examined the feasibility and efficacy of skeletal muscle injection of adeno-associated virus 5 encoding IGF-I under Tet regulation (AAV5.IGFI-tet) to treat heart failure. Myocardial infarction (MI) was induced in rats by coronary occlusion; 1 week later, rats with impaired left ventricular (LV) function received 2×10(12) genome copies (GC) of AAV5.IGFI-tet in the anterior tibialis muscle, and 4 weeks later, were randomly assigned to receive doxycycline in drinking water to activate IGF-I expression (IGF-On; n=10), or not to receive doxycycline (IGF-Off; n=10). Ten weeks after MI (5 weeks after activation of IGF-I expression), LV size and function were assessed by echocardiography and physiological studies. IGF-On rats showed reduced LV end-systolic dimension (p=0.03) and increased LV ejection fraction (p=0.02). In addition, IGF-On rats showed, before and during dobutamine infusion, increases in cardiac output (p=0.02), stroke work (p=0.0001), LV + dP/dt (p<0.0001), LV relaxation (LV - dP/dt; p=0.03), and systolic arterial blood pressure (p=0.0003). Mean arterial pressure and systemic vascular resistance were unchanged. Activation of IGF-I expression reduced cardiac fibrosis (p=0.048), apoptosis (p<0.0001), and caspase-3/7 activity (p=0.04). Serum IGF-I was increased 5 weeks after transgene activation (p=0.008). These data indicate that skeletal muscle injection of AAV5.IGFI-tet enables tetracycline-activated expression, increases serum IGF-I levels, and improves function of the failing heart.
Collapse
Affiliation(s)
- N Chin Lai
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | | | | | | | | | | |
Collapse
|
28
|
A mouse model for spatial and temporal expression of HGF in the heart. Transgenic Res 2011; 20:1203-16. [DOI: 10.1007/s11248-011-9485-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 01/09/2011] [Indexed: 10/18/2022]
|
29
|
Activated Met signalling in the developing mouse heart leads to cardiac disease. PLoS One 2011; 6:e14675. [PMID: 21347410 PMCID: PMC3036588 DOI: 10.1371/journal.pone.0014675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 01/13/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The Hepatocyte Growth Factor (HGF) is a pleiotropic cytokine involved in many physiological processes, including skeletal muscle, placenta and liver development. Little is known about its role and that of Met tyrosine kinase receptor in cardiac development. METHODOLOGY/PRINCIPAL FINDINGS In this study, we generated two transgenic mice with cardiac-specific, tetracycline-suppressible expression of either Hepatocyte Growth Factor (HGF) or the constitutively activated Tpr-Met kinase to explore: i) the effect of stimulation of the endogenous Met receptor by autocrine production of HGF and ii) the consequence of sustained activation of Met signalling in the heart. We first showed that Met is present in the neonatal cardiomyocytes and is responsive to exogenous HGF. Exogenous HGF starting from prenatal stage enhanced cardiac proliferation and reduced sarcomeric proteins and Connexin43 (Cx43) in newborn mice. As adults, these transgenics developed systolic contractile dysfunction. Conversely, prenatal Tpr-Met expression was lethal after birth. Inducing Tpr-Met expression during postnatal life caused early-onset heart failure, characterized by decreased Cx43, upregulation of fetal genes and hypertrophy. CONCLUSIONS/SIGNIFICANCE Taken together, our data show that excessive activation of the HGF/Met system in development may result in cardiac damage and suggest that Met signalling may be implicated in the pathogenesis of cardiac disease.
Collapse
|
30
|
Lu S, Crawford GL, Dore J, Anderson SA, Despres D, Horowits R. Cardiac-specific NRAP overexpression causes right ventricular dysfunction in mice. Exp Cell Res 2011; 317:1226-37. [PMID: 21276443 DOI: 10.1016/j.yexcr.2011.01.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/14/2011] [Accepted: 01/19/2011] [Indexed: 01/08/2023]
Abstract
The muscle-specific protein NRAP is concentrated at cardiac intercalated disks, plays a role in myofibril assembly, and is upregulated early in mouse models of dilated cardiomyopathy. Using a tet-off system, we developed novel transgenic lines exhibiting cardiac-specific NRAP overexpression ~2.5 times greater than normal. At 40-50 weeks, NRAP overexpression resulted in dilation and decreased ejection fraction in the right ventricle, with little effect on the left ventricle. Expression of transcripts encoding brain natriuretic peptide and skeletal α-actin was increased by cardiac-specific NRAP overexpression, indicative of a cardiomyopathic response. NRAP overexpression did not alter the levels or organization of N-cadherin and connexin-43. The results show that chronic NRAP overexpression in the mouse leads to right ventricular cardiomyopathy by 10 months, but that the early NRAP upregulation previously observed in some mouse models of dilated cardiomyopathy is unlikely to account for the remodeling of intercalated disks and left ventricular dysfunction observed in those cases.
Collapse
Affiliation(s)
- Shajia Lu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
31
|
Caruso L, Yuen S, Smith J, Husain M, Opavsky MA. Cardiomyocyte-targeted overexpression of the coxsackie–adenovirus receptor causes a cardiomyopathy in association with β-catenin signaling. J Mol Cell Cardiol 2010; 48:1194-205. [DOI: 10.1016/j.yjmcc.2010.01.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 12/29/2009] [Accepted: 01/30/2010] [Indexed: 01/09/2023]
|
32
|
Wu B, Zhou B, Wang Y, Cheng HL, Hang CT, Pu WT, Chang CP, Zhou B. Inducible cardiomyocyte-specific gene disruption directed by the rat Tnnt2 promoter in the mouse. Genesis 2010; 48:63-72. [PMID: 20014345 DOI: 10.1002/dvg.20573] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We developed a conditional and inducible gene knockout methodology that allows effective gene deletion in mouse cardiomyocytes. This transgenic mouse line was generated by coinjection of two transgenes, a "reverse" tetracycline-controlled transactivator (rtTA) directed by a rat cardiac troponin T (Tnnt2) promoter and a Cre recombinase driven by a tetracycline-responsive promoter (TetO). Here, Tnnt2-rtTA activated TetO-Cre expression takes place in cardiomyocytes following doxycycline treatment. Using two different mouse Cre reporter lines, we demonstrated that expression of Cre recombinase was specifically and robustly induced in the cardiomyocytes of embryonic or adult hearts following doxycycline induction, thus, allowing cardiomyocyte-specific gene disruption and lineage tracing. We also showed that rtTA expression and doxycycline treatment did not compromise cardiac function. These features make the Tnnt2-rtTA;TetO-Cre transgenic line a valuable genetic tool for analysis of spatiotemporal gene function and cardiomyocyte lineage tracing during developmental and postnatal periods.
Collapse
Affiliation(s)
- Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Price Center 420, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
"Pacemaker" f-channels mediating the hyperpolarization-activated nonselective cation current I(f) are directly regulated by cAMP. Accordingly, the activity of f-channels increases when cellular cAMP levels are elevated (e.g., during sympathetic stimulation) and decreases when they are reduced (e.g., during vagal stimulation). Although these biophysical properties seem to make f-channels ideal molecular targets for heart rate regulation by the autonomic nervous system, the exact contribution of the major I(f)-mediating cardiac isoforms HCN2 and HCN4 to sinoatrial node (SAN) function remains highly controversial. To directly investigate the role of cAMP-dependent regulation of hyperpolarization activated cyclic nucleotide activated (HCN) channels in SAN activity, we generated mice with heart-specific and inducible expression of a human HCN4 mutation (573X) that abolishes the cAMP-dependent regulation of HCN channels. We found that hHCN4-573X expression causes elimination of the cAMP sensitivity of I(f) and decreases the maximum firing rates of SAN pacemaker cells. In conscious mice, hHCN4-573X expression leads to a marked reduction in heart rate at rest and during exercise. Despite the complete loss of cAMP sensitivity of I(f), the relative extent of SAN cell frequency and heart rate regulation are preserved. Our data demonstrate that cAMP-mediated regulation of I(f) determines basal and maximal heart rates but does not play an indispensable role in heart rate adaptation during physical activity. Our data also reveal the pathophysiologic mechanism of hHCN4-573X-linked SAN dysfunction in humans.
Collapse
|
34
|
Palais G, Nguyen Dinh Cat A, Friedman H, Panek-Huet N, Millet A, Tronche F, Gellen B, Mercadier JJ, Peterson A, Jaisser F. Targeted transgenesis at the HPRT locus: an efficient strategy to achieve tightly controlled in vivo conditional expression with the tet system. Physiol Genomics 2009; 37:140-6. [DOI: 10.1152/physiolgenomics.90328.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tet-inducible system has been widely used to achieve conditional gene expression in genetically modified mice. To alleviate the frequent difficulties associated with recovery of relevant transgenic founders, we tested whether a controlled strategy of transgenesis would support reliable cell-specific, doxycycline (Dox)-controlled transgene expression in vivo. Taking advantage of the potent hypoxanthine-aminopterin-thymidine selection strategy and an embryonic stem (ES) cell line supporting efficient germ-line transmission, we used hypoxanthine phosphoribosyltransferase ( HPRT) targeting to insert a single copy tet-inducible construct designed to allow both glucocorticoid receptor (GR) and β-galactosidase (β-Gal) expression. Conditional, Dox-dependent GR and β-Gal expression was evidenced in targeted ES cells. Breeding ES-derived single copy transgenic mice with mice bearing appropriate tet transactivators resulted in β-Gal expression both qualitatively and quantitatively similar to that observed in mice with random integration of the same construct. Interestingly, GR expression in mice was dependent on transgene orientation in the HPRT locus while embryonic stem cell expression was not. Thus, a conditional construct inserted in single copy and in predetermined orientation at the HPRT locus demonstrated a Dox-dependent gene expression phenotype in adult mice suggesting that controlled insertion of tet-inducible constructs at the HPRT locus can provide an efficient alternative strategy to reproducibly generate animal models with tetracycline-induced transgene expression.
Collapse
Affiliation(s)
- G. Palais
- Institut National de la Santé et de la Recherche Médicale (INSERM), U772
- Collège de France
- l'Université Paris Descartes, Paris, France
| | - A. Nguyen Dinh Cat
- Institut National de la Santé et de la Recherche Médicale (INSERM), U772
- Collège de France
- l'Université Paris Descartes, Paris, France
| | - H. Friedman
- Laboratory of Developmental Biology, H-5, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - N. Panek-Huet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U772
- Collège de France
- l'Université Paris Descartes, Paris, France
| | - A. Millet
- Collège de France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 7148
| | - F. Tronche
- Collège de France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 7148
| | - B. Gellen
- INSERM, U698
- l'Université Paris 7, Paris, France
| | | | - A. Peterson
- Laboratory of Developmental Biology, H-5, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - F. Jaisser
- Institut National de la Santé et de la Recherche Médicale (INSERM), U772
- Collège de France
- l'Université Paris Descartes, Paris, France
| |
Collapse
|
35
|
Ueno S, Pease ME, Wersinger DMB, Masuda T, Vinores SA, Licht T, Zack DJ, Quigley H, Keshet E, Campochiaro PA. Prolonged blockade of VEGF family members does not cause identifiable damage to retinal neurons or vessels. J Cell Physiol 2008; 217:13-22. [PMID: 18543272 DOI: 10.1002/jcp.21445] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Several ocular diseases complicated by neovascularization are being treated by repeated intraocular injections of vascular endothelial growth factor (VEGF) antagonists. While substantial benefits have been documented, there is concern that unrecognized damage may be occurring, because blockade of VEGF may damage the fenestrated vessels of the choroicapillaris and deprive retinal neurons of input from a survival factor. One report has suggested that even temporary blockade of all isoforms of VEGF-A results in significant loss of retinal ganglion cells. In this study, we utilized double transgenic mice with doxycycline-inducible expression of soluble VEGF receptor 1 coupled to an Fc fragment (sVEGFR1Fc), a potent antagonist of several VEGF family members, including VEGF-A, to test the effects of VEGF blockade in the retina. Expression of sVEGFR1Fc completely blocked VEGF-induced retinal vascular permeability and significantly suppressed the development of choroidal neovascularization at rupture sites in Bruch's membrane, but did not cause regression of established choroidal neovascularization. Mice with constant expression of sVEGFR1Fc in the retina for 7 months had normal electroretinograms and normal retinal and choroidal ultrastructure including normal fenestrations in the choroicapillaris. They also showed no significant difference from control mice in the number of ganglion cell axons in optic nerve cross sections and the retinal level of mRNA for 3 ganglion cell-specific genes. These data indicate that constant blockade of VEGF for up to 7 months has no identifiable deleterious effects on the retina or choroid and support the use of VEGF antagonists in the treatment of retinal diseases.
Collapse
Affiliation(s)
- Shinji Ueno
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Vinet L, Rouet-Benzineb P, Marniquet X, Pellegrin N, Mangin L, Louedec L, Samuel JL, Mercadier JJ. Chronic doxycycline exposure accelerates left ventricular hypertrophy and progression to heart failure in mice after thoracic aorta constriction. Am J Physiol Heart Circ Physiol 2008; 295:H352-60. [PMID: 18487442 DOI: 10.1152/ajpheart.01101.2007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tetracycline is a powerful tool for controlling the expression of specific transgenes (TGs) in various tissues, including heart. In these mouse systems, TG expression is repressed/enhanced by adding doxycycline (Dox) to the diet. However, Dox has been shown to attenuate matrix metalloproteinase (MMP) expression and activity in various tissues, and MMP inactivation mitigates left ventricular (LV) remodeling in animal models of heart failure. Therefore, we examined the influence of Dox on LV remodeling and MMP expression in mice after transverse aortic constriction (TAC). One month after TAC, cardiac hypertrophy (99% vs. 67%) and the proportion of mice exhibiting congestive heart failure (CHF, 74% vs. 32%) were higher in the TAC + Dox group than in the TAC group (P < 0.05). These differences were no longer seen 2 mo after TAC, although LV was more severely dilated in TAC + Dox mice than in TAC mice (P < 0.05). One month after TAC, the increase in brain natriuretic peptide and beta-myosin heavy chain mRNA levels was 1.6 and 1.7 times higher, respectively, in TAC + Dox mice than in TAC mice (P < 0.01). MMP-2 gelatin zymographic activity increased 1.9- and 2.4-fold in TAC and TAC + Dox mice, respectively (P < 0.01 and P < 0.05 relative to respective sham-operated animals), but the difference between TAC + Dox and TAC mice did not reach statistical significance. Dox did not significantly alter TAC-associated perivascular and interstitial myocardial fibrosis. These findings demonstrate that Dox accelerates the onset of cardiac hypertrophy and the progression to CHF following TAC in mice. Accordingly, care should be taken when designing and interpreting studies based on TG mouse models of LV hypertrophy using the tetracycline-regulated (tet)-on/tet-off system.
Collapse
Affiliation(s)
- Laurent Vinet
- INSERM U698, G. H. Bichat-Claude Bernard, 75877 Paris Cedex 18, France.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Tirziu D, Chorianopoulos E, Moodie KL, Palac RT, Zhuang ZW, Tjwa M, Roncal C, Eriksson U, Fu Q, Elfenbein A, Hall AE, Carmeliet P, Moons L, Simons M. Myocardial hypertrophy in the absence of external stimuli is induced by angiogenesis in mice. J Clin Invest 2008; 117:3188-97. [PMID: 17975666 DOI: 10.1172/jci32024] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Accepted: 08/29/2007] [Indexed: 01/09/2023] Open
Abstract
Although studies have suggested a role for angiogenesis in determining heart size during conditions demanding enhanced cardiac performance, the role of EC mass in determining the normal organ size is poorly understood. To explore the relationship between cardiac vasculature and normal heart size, we generated a transgenic mouse with a regulatable expression of the secreted angiogenic growth factor PR39 in cardiomyocytes. A significant change in adult mouse EC mass was apparent by 3 weeks following PR39 induction. Heart weight; cardiomyocyte size; vascular density normalization; upregulation of hypertrophy markers including atrial natriuretic factor, beta-MHC, and GATA4; and activation of the Akt and MAP kinase pathways were observed at 6 weeks post-induction. Treatment of PR39-induced mice with the eNOS inhibitor L-NAME in the last 3 weeks of a 6-week stimulation period resulted in a significant suppression of heart growth and a reduction in hypertrophic marker expression. Injection of PR39 or another angiogenic growth factor, VEGF-B, into murine hearts during myocardial infarction led to induction of myocardial hypertrophy and restoration of myocardial function. Thus stimulation of vascular growth in normal adult mouse hearts leads to an increase in cardiac mass.
Collapse
Affiliation(s)
- Daniela Tirziu
- Angiogenesis Research Center, Section of Cardiology, Department of Medicine, Dartmouth Medical School, Hanover, New Hampshire 03756, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Transgenic system for conditional induction and rescue of chronic myocardial hibernation provides insights into genomic programs of hibernation. Proc Natl Acad Sci U S A 2007; 105:282-7. [PMID: 18162550 DOI: 10.1073/pnas.0707778105] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
A key energy-saving adaptation to chronic hypoxia that enables cardiomyocytes to withstand severe ischemic insults is hibernation, i.e., a reversible arrest of contractile function. Whereas hibernating cardiomyocytes represent the critical reserve of dysfunctional cells that can be potentially rescued, a lack of a suitable animal model has hampered insights on this medically important condition. We developed a transgenic mouse system for conditional induction of long-term hibernation and a system to rescue hibernating cardiomyocytes at will. Via myocardium-specific induction (and, in turn, deinduction) of a VEGF-sequestering soluble receptor, we show that VEGF is indispensable for adjusting the coronary vasculature to match increased oxygen consumption and exploit this finding to generate a hypoperfused heart. Importantly, ensuing ischemia is tunable to a level at which large cohorts of cardiomyocytes are driven to enter a hibernation mode, without cardiac cell death. Relieving the VEGF blockade even months later resulted in rapid revascularization and full recovery of contractile function. Furthermore, we show that left ventricular remodeling associated with hibernation is also fully reversible. The unique opportunity to uncouple hibernation from other ischemic heart phenotypes (e.g., infarction) was used to determine the genetic program of hibernation; uncovering hypoxia-inducible factor target genes associated with metabolic adjustments and induced expression of several cardioprotective genes. Autophagy, specifically self-digestion of mitochondria, was identified as a key prosurvival mechanism in hibernating cardiomyocytes. This system may lend itself for examining the potential utility of treatments to rescue dysfunctional cardiomyocytes and reverse maladaptive remodeling.
Collapse
|
39
|
|
40
|
McCloskey DT, Turcato S, Wang GY, Turnbull L, Zhu BQ, Bambino T, Nguyen AP, Lovett DH, Nissenson RA, Karliner JS, Baker AJ. Expression of a Gi-coupled receptor in the heart causes impaired Ca2+ handling, myofilament injury, and dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2007; 294:H205-12. [PMID: 17965283 DOI: 10.1152/ajpheart.00829.2007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Increased signaling by G(i)-coupled receptors has been implicated in dilated cardiomyopathy. To investigate the mechanisms, we used transgenic mice that develop dilated cardiomyopathy after conditional expression of a cardiac-targeted G(i)-coupled receptor (Ro1). Activation of G(i) signaling by the Ro1 agonist spiradoline caused decreased cellular cAMP levels and bradycardia in Langendorff-perfused hearts. However, acute termination of Ro1 signaling with the antagonist nor-binaltorphimine did not reverse the Ro1-induced contractile dysfunction, indicating that Ro1 cardiomyopathy was not due to acute effects of receptor signaling. Early after initiation of Ro1 expression, there was a 40% reduction in the abundance of the sarcoplasmic reticulum Ca(2+)-ATPase (P < 0.05); thereafter, there was progressive impairment of both Ca(2+) handling and force development assessed with ventricular trabeculae. Six weeks after initiation of Ro1 expression, systolic Ca(2+) concentration was reduced to 0.61 +/- 0.08 vs. 0.91 +/- 0.07 microM for control (n = 6-8; P < 0.05), diastolic Ca(2+) concentration was elevated to 0.41 +/- 0.07 vs. 0.23 +/- 0.06 microM for control (n = 6-8; P < 0.01), and the decline phase of the Ca(2+) transient (time from peak to 50% decline) was slowed to 0.25 +/- 0.02 s vs. 0.13 +/- 0.02 s for control (n = 6-8; P < 0.01). Early after initiation of Ro1 expression, there was a ninefold elevation of matrix metalloproteinase-2 (P < 0.01), which is known to cause myofilament injury. Consistent with this, 6 wk after initiation of Ro1 expression, Ca(2+)-saturated myofilament force in skinned trabeculae was reduced to 21 +/- 2 vs. 38 +/- 0.1 mN/mm(2) for controls (n = 3; P < 0.01). Furthermore, electron micrographs revealed extensive myofilament damage. These findings may have implications for some forms of human heart failure in which increased activity of G(i)-coupled receptors leads to impaired Ca(2+) handling and myofilament injury, contributing to impaired ventricular pump function and heart failure.
Collapse
Affiliation(s)
- Diana T McCloskey
- VA Medical Center, Cardiology Division (111C ), University of California-San Francisco, 4150 Clement St., San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ernst E, Schönig K, Bugert JJ, Bläker H, Pfaff E, Stremmel W, Encke J. Generation of inducible hepatitis C virus transgenic mouse lines. J Med Virol 2007; 79:1103-12. [PMID: 17596832 DOI: 10.1002/jmv.20911] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Hepatitis C virus (HCV) is the causative agent of most cases of chronic liver disease, cirrhosis, and hepatocellular carcinoma (HCC) affecting more than 170 million people world-wide. Progress in elucidating the nature of HCV and the development of new therapeutic strategies is hampered fundamentally by the absence of adequate small animal models simulating natural HCV infection. The creation of conditional mouse lines with the tetracycline-controlled gene expression system holds new perspectives for simulation of wild-type HCV infection in a small animal model. Transgenic mice were established with tetracycline-inducible coexpression of HCV core or HCV open reading frame (ORF) and luciferase. In long-term induction experiments, mice were examined for immunopathological changes after expression of HCV proteins. Inducible and liver-specific expression of transgenes was detected by Western blot, immunoprecipitation, luciferase assay and in vivo imaging of bioluminescence of luciferase in genetically modified mice. Ectopic expression levels were determined quantitatively in the liver, kidney, heart and spleen of mice in the induced and non-induced state. During long-term induction an elevation of aminotransaminases (ALT) was observed only in HCV core/ORF-expressing mice, but HCV-specific immune response was not confirmed by in vitro immunological assays. The histology of liver sections provided evidence of steatosis, which was correlated with an inflammatory response. The inducible HCV-transgenic mouse lines provide further evidence of liver pathogenesis in the presence of inflammation during liver-specific expression of HCV proteins and offer new insights into the effects of temporally and spatially controlled protein expression of HCV.
Collapse
Affiliation(s)
- Evelyn Ernst
- Internal Medicine IV, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
42
|
Sainte-Marie Y, Nguyen Dinh Cat A, Perrier R, Mangin L, Soukaseum C, Peuchmaur M, Tronche F, Farman N, Escoubet B, Benitah JP, Jaisser F. Conditional glucocorticoid receptor expression in the heart induces atrio-ventricular block. FASEB J 2007; 21:3133-41. [PMID: 17517920 DOI: 10.1096/fj.07-8357com] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Corticosteroid hormones (aldosterone and glucocorticoids) and their receptors are now recognized as major modulators of cardiovascular pathophysiology, but their specific roles remain elusive. Glucocorticoid hormones (GCs), which are widely used to treat acute and chronic diseases, often have adverse cardiovascular effects such as heart failure, hypertension, atherosclerosis, or metabolic alterations. The direct effects of GC on the heart are difficult to evaluate, as changes in plasma GC concentrations have multiple consequences due to the ubiquitous expression of the glucocorticoid receptor (GR), resulting in secondary effects on cardiac function. We evaluated the effects of GR on the heart in a conditional mouse model in which the GR was overexpressed solely in cardiomyocytes. The transgenic mice displayed electrocardiogram (ECG) abnormalities: a long PQ interval, increased QRS and QTc duration as well as chronic atrio-ventricular block, without cardiac hypertrophy or fibrosis. The ECG alterations were reversible on GR expression shutoff. Isolated ventricular cardiomyocytes showed major ion channel remodeling, with decreases in I(Na), I(to), and I(Kslow) activity and changes in cell calcium homeostasis (increase in C(al), in Ca2+ transients and in sarcoplasmic reticulum Ca2+ load). This phenotype differs from that observed in mice overexpressing the mineralocorticoid receptor in the heart, which displayed ventricular arrhythmia. Our mouse model highlights novel effects of GR activation in the heart indicating that GR has direct and specific cardiac effects in the mouse.
Collapse
|
43
|
Nguyen Din Cat A, Sainte-Marie Y, Jaisser F. Animal models in cardiovascular diseases: new insights from conditional models. Handb Exp Pharmacol 2007:377-405. [PMID: 17203664 DOI: 10.1007/978-3-540-35109-2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conditional systems have proven to be efficient and powerful to delineate several aspects of cardiac pathophysiology and diseases. The possibility of addressing a particular time point in animal life is certainly an important breakthrough allowed by conditional strategies with temporal control of either transgene expression or gene modifications. The purpose of this review is to present various mouse models for cardiovascular diseases based on conditional approaches.
Collapse
Affiliation(s)
- A Nguyen Din Cat
- INSERM U772, College De France, 11 Place Marcelin Berthelot, 75231 Parisx 05, France
| | | | | |
Collapse
|
44
|
Trivieri MG, Oudit GY, Sah R, Kerfant BG, Sun H, Gramolini AO, Pan Y, Wickenden AD, Croteau W, Morreale de Escobar G, Pekhletski R, St. Germain D, MacLennan DH, Backx PH. Cardiac-specific elevations in thyroid hormone enhance contractility and prevent pressure overload-induced cardiac dysfunction. Proc Natl Acad Sci U S A 2006; 103:6043-8. [PMID: 16595628 PMCID: PMC1426242 DOI: 10.1073/pnas.0601072103] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Thyroid hormone (TH) is critical for cardiac development and heart function. In heart disease, TH metabolism is abnormal, and many biochemical and functional alterations mirror hypothyroidism. Although TH therapy has been advocated for treating heart disease, a clear benefit of TH has yet to be established, possibly because of peripheral actions of TH. To assess the potential efficacy of TH in treating heart disease, type 2 deiodinase (D2), which converts the prohormone thyroxine to active triiodothyronine (T3), was expressed transiently in mouse hearts by using the tetracycline transactivator system. Increased cardiac D2 activity led to elevated cardiac T3 levels and to enhanced myocardial contractility, accompanied by increased Ca(2+) transients and sarcoplasmic reticulum (SR) Ca(2+) uptake. These phenotypic changes were associated with up-regulation of sarco(endo)plasmic reticulum calcium ATPase (SERCA) 2a expression as well as decreased Na(+)/Ca(2+) exchanger, beta-myosin heavy chain, and sarcolipin (SLN) expression. In pressure overload, targeted increases in D2 activity could not block hypertrophy but could completely prevent impaired contractility and SR Ca(2+) cycling as well as altered expression patterns of SERCA2a, SLN, and other markers of pathological hypertrophy. Our results establish that elevated D2 activity in the heart increases T3 levels and enhances cardiac contractile function while preventing deterioration of cardiac function and altered gene expression after pressure overload.
Collapse
Affiliation(s)
- Maria Giovanna Trivieri
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Gavin Y. Oudit
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Rajan Sah
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Benoit-Gilles Kerfant
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Hui Sun
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Anthony O. Gramolini
- Banting and Best Department of Medical Research, Charles H. Best Institute, University of Toronto, Toronto, ON, Canada M5G 1L6
| | - Yan Pan
- Banting and Best Department of Medical Research, Charles H. Best Institute, University of Toronto, Toronto, ON, Canada M5G 1L6
| | - Alan D. Wickenden
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Walburga Croteau
- Department of Medicine, Dartmouth Medical School, Lebanon, NH 03756; and
| | | | - Roman Pekhletski
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| | - Donald St. Germain
- Department of Medicine, Dartmouth Medical School, Lebanon, NH 03756; and
| | - David H. MacLennan
- Banting and Best Department of Medical Research, Charles H. Best Institute, University of Toronto, Toronto, ON, Canada M5G 1L6
| | - Peter H. Backx
- *Heart and Stroke/Richard Lewar Centre and Departments of Physiology and Medicine, University of Toronto, Toronto, ON, Canada M5S 3E2
- Division of Cardiology, University Health Network, Toronto, ON, Canada M5S 3E2
| |
Collapse
|
45
|
Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Jung S, Yung S, Chimenti S, Landsman L, Abramovitch R, Keshet E. VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 2006; 124:175-89. [PMID: 16413490 DOI: 10.1016/j.cell.2005.10.036] [Citation(s) in RCA: 895] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2004] [Revised: 08/11/2005] [Accepted: 10/07/2005] [Indexed: 01/31/2023]
Abstract
Adult neovascularization relies on the recruitment of circulating cells, but their angiogenic roles and recruitment mechanisms are unclear. We show that the endothelial growth factor VEGF is sufficient for organ homing of circulating mononuclear myeloid cells and is required for their perivascular positioning and retention. Recruited bone marrow-derived circulating cells (RBCCs) summoned by VEGF serve a function distinct from endothelial progenitor cells. Retention of RBCCs in close proximity to angiogenic vessels is mediated by SDF1, a chemokine induced by VEGF in activated perivascular myofibroblasts. RBCCs enhance in situ proliferation of endothelial cells via secreting proangiogenic activities distinct from locally induced activities. Precluding RBCCs strongly attenuated the proangiogenic response to VEGF and addition of purified RBCCs enhanced angiogenesis in excision wounds. Together, the data suggest a model for VEGF-programmed adult neovascularization highlighting the essential paracrine role of recruited myeloid cells and a role for SDF1 in their perivascular retention.
Collapse
Affiliation(s)
- Myriam Grunewald
- Department of Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Turnbull L, Zhou HZ, Swigart PM, Turcato S, Karliner JS, Conklin BR, Simpson PC, Baker AJ. Sustained preconditioning induced by cardiac transgenesis with the tetracycline transactivator. Am J Physiol Heart Circ Physiol 2006; 290:H1103-9. [PMID: 16243914 DOI: 10.1152/ajpheart.00732.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Preconditioning protocols that protect the heart from ischemic injury may aid in the development of new therapies. However, the temporal window of cardioprotection is limited to a few days after the preconditioning stimulus. Here we report a sustained cardioprotected phenotype in mice expressing a tetracycline transactivator (tTA) transcription factor under the control of the alpha-myosin heavy chain (alphaMHC) promoter. alphaMHC-tTA mice were originally designed for tetracycline-regulated gene expression in the heart (Tet system). However, we found that after 45 min of global ischemia at 37 degrees C, left ventricular developed pressure (LVDP) of Langendorff-perfused alphaMHC-tTA mouse hearts rapidly recovered in 5 min to 60% of initial levels, whereas LVDP of wild-type (WT) littermates recovered to only 10% of the initial level. Improved postischemic recovery of function for alphaMHC-tTA hearts was associated with a 50% decrease of infarct size and a significantly smaller release of lactate dehydrogenase to the coronary effluent. Improved postischemic recovery was not attributable to differences in coronary flow that was similar for WT- and alphaMHC-tTA hearts during recovery. Moreover, improved postischemic recovery of alphaMHC-tTA hearts was not abolished by inhibitors of classical cardioprotective effectors (mitochondrial ATP-sensitive K+ channels, PKC, or adenosine receptors), suggesting a novel mechanism. Finally, the tetracycline analog doxycycline, which inhibits binding of tTA to DNA, did not abolish improved recovery for alphaMHC-tTA hearts. The sustained cardioprotected phenotype of alphaMHC-tTA hearts may have implications for developing new therapies to minimize cardiac ischemic injury. Furthermore, investigations of cardioprotection using the Tet system may be aberrantly influenced by sustained preconditioning induced by cardiac transgenesis with tTA.
Collapse
Affiliation(s)
- Lynne Turnbull
- Department of Radiology, University of California, San Francisco, California 94121, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee S, Agah R, Xiao M, Frutkin AD, Kremen M, Shi H, Dichek DA. In vivo expression of a conditional TGF-beta1 transgene: no evidence for TGF-beta1 transgene expression in SM22alpha-tTA transgenic mice. J Mol Cell Cardiol 2005; 40:148-56. [PMID: 16288910 PMCID: PMC1444940 DOI: 10.1016/j.yjmcc.2005.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2005] [Revised: 09/20/2005] [Accepted: 09/26/2005] [Indexed: 11/28/2022]
Abstract
Transforming growth beta-1 (TGF-beta1) appears to play a critical role in the regulation of arterial intimal growth and the development of atherosclerosis. TGF-beta1 is expressed at increased levels in diseased arteries; however, its role in disease development remains controversial. Experiments in which TGF-beta1 is overexpressed in the artery wall of transgenic mice could clarify the role of TGF-beta1 in the development or prevention of vascular disease. However, constitutive overexpression of a TGF-beta1 transgene in the mouse artery wall is embryonically lethal. Therefore, to overexpress TGF-beta1 in the artery wall of adult mice, we generated mice that were transgenic for a conditional, tetracycline operator (tetO)-driven TGF-beta1 allele. These mice were viable, and when crossed with mice expressing a tetracycline-regulated transactivator (tTA) in the heart, expressed the TGF-beta1 transgene in a cardiac-restricted and doxycycline-dependent manner. Nevertheless, breeding of the tetO-TGF-beta1 transgene into three lines of mice transgenic for a smooth muscle-targeted tTA (SM22alpha-tTA mice; reported elsewhere to transactivate tetO-driven alleles in smooth muscle cells of large arteries) did not yield expression of the TGF-beta1 transgene. Moreover, tTA expression was not detected in aortae of the SM22alpha-tTA mice. Transgenic mice that express tTA at high levels in vascular smooth muscle and reliably transactivate tetO-driven transgenes would be useful for deciphering the role of TGF-beta1 (or other proteins) in normal arterial physiology and in the development of arterial disease. Currently available SM22alpha-tTA mice were not useful for this purpose. Generation of higher-expressing lines of SM22alpha-tTA mice appears warranted.
Collapse
Affiliation(s)
- Sunyoung Lee
- Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Shiojima I, Sato K, Izumiya Y, Schiekofer S, Ito M, Liao R, Colucci WS, Walsh K. Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. J Clin Invest 2005; 115:2108-18. [PMID: 16075055 PMCID: PMC1180541 DOI: 10.1172/jci24682] [Citation(s) in RCA: 757] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Accepted: 05/17/2005] [Indexed: 12/18/2022] Open
Abstract
Although increased external load initially induces cardiac hypertrophy with preserved contractility, sustained overload eventually leads to heart failure through poorly understood mechanisms. Here we describe a conditional transgenic system in mice characterized by the sequential development of adaptive cardiac hypertrophy with preserved contractility in the acute phase and dilated cardiomyopathy in the chronic phase following the induction of an activated Akt1 gene in the heart. Coronary angiogenesis was enhanced during the acute phase of adaptive cardiac growth but reduced as hearts underwent pathological remodeling. Enhanced angiogenesis in the acute phase was associated with mammalian target of rapamycin-dependent induction of myocardial VEGF and angiopoietin-2 expression. Inhibition of angiogenesis by a decoy VEGF receptor in the acute phase led to decreased capillary density, contractile dysfunction, and impaired cardiac growth. Thus, both heart size and cardiac function are angiogenesis dependent, and disruption of coordinated tissue growth and angiogenesis in the heart contributes to the progression from adaptive cardiac hypertrophy to heart failure.
Collapse
Affiliation(s)
- Ichiro Shiojima
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
McCloskey DT, Turnbull L, Swigart PM, Zambon AC, Turcato S, Joho S, Grossman W, Conklin BR, Simpson PC, Baker AJ. Cardiac transgenesis with the tetracycline transactivator changes myocardial function and gene expression. Physiol Genomics 2005; 22:118-26. [PMID: 15797971 DOI: 10.1152/physiolgenomics.00016.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cardiac-specific tetracycline-regulated gene expression system (tet-system) is a powerful tool using double-transgenic mice. The cardiac alpha-myosin heavy chain promoter (alphaMHC) drives lifetime expression of a tetracycline-inhibited transcription activator (tTA). Crossing alphaMHC-tTA mice with mice containing a tTA-responsive promoter linked to a target gene yields double-transgenic mice having tetracycline-repressed expression of the target gene in the heart. Using the tet-system, some studies use nontransgenic mice for the control group, whereas others use single-transgenic alphaMHC-tTA mice. However, previous studies found that high-level expression of a modified activator protein caused cardiomyopathy. Therefore, we tested whether cardiac expression of tTA was associated with altered function of alphaMHC-tTA mice compared with wild-type (WT) littermates. We monitored in vivo and in vitro function and gene expression profiles for myocardium from WT and alphaMHC-tTA mice. Compared with WT littermates, alphaMHC-tTA mice had a greater heart-to-body weight ratio (approximately 10%), ventricular dilation, and decreased ejection fraction, suggesting mild cardiomyopathy. In vitro, submaximal contractions were greater compared with WT and were associated with greater myofilament Ca2+ sensitivity. Gene expression profiling revealed that the expression of 153 genes was significantly changed by >20% when comparing alphaMHC-tTA with WT myocardium. These findings demonstrate that introduction of the alphaMHC-tTA construct causes significant effects on myocardial gene expression and major functional abnormalities in vivo and in vitro. For studies using the tet-system, these results suggest caution in the use of controls, since alphaMHC-tTA myocardium differs appreciably from WT. Furthermore, the results raise the possibility that the phenotype conferred by a target gene may be influenced by the modified genetic background of alphaMHC-tTA myocardium.
Collapse
Affiliation(s)
- Diana T McCloskey
- Department of Radiology, University of California, San Francisco, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ouvrard-Pascaud A, Sainte-Marie Y, Bénitah JP, Perrier R, Soukaseum C, Nguyen Dinh Cat A, Royer A, Le Quang K, Charpentier F, Demolombe S, Mechta-Grigoriou F, Beggah AT, Maison-Blanche P, Oblin ME, Delcayre C, Fishman GI, Farman N, Escoubet B, Jaisser F. Conditional mineralocorticoid receptor expression in the heart leads to life-threatening arrhythmias. Circulation 2005; 111:3025-33. [PMID: 15939817 PMCID: PMC3635833 DOI: 10.1161/circulationaha.104.503706] [Citation(s) in RCA: 202] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Life-threatening cardiac arrhythmia is a major source of mortality worldwide. Besides rare inherited monogenic diseases such as long-QT or Brugada syndromes, which reflect abnormalities in ion fluxes across cardiac ion channels as a final common pathway, arrhythmias are most frequently acquired and associated with heart disease. The mineralocorticoid hormone aldosterone is an important contributor to morbidity and mortality in heart failure, but its mechanisms of action are incompletely understood. METHODS AND RESULTS To specifically assess the role of the mineralocorticoid receptor (MR) in the heart, in the absence of changes in aldosteronemia, we generated a transgenic mouse model with conditional cardiac-specific overexpression of the human MR. Mice exhibit a high rate of death prevented by spironolactone, an MR antagonist used in human therapy. Cardiac MR overexpression led to ion channel remodeling, resulting in prolonged ventricular repolarization at both the cellular and integrated levels and in severe ventricular arrhythmias. CONCLUSIONS Our results indicate that cardiac MR triggers cardiac arrhythmias, suggesting novel opportunities for prevention of arrhythmia-related sudden death.
Collapse
|