1
|
Li Q, Homilius M, Achilles E, Massey LK, Convey V, Ohlsson Å, Ljungvall I, Häggström J, Boler BV, Steiner P, Day S, MacRae CA, Oyama MA. Metabolic abnormalities and reprogramming in cats with naturally occurring hypertrophic cardiomyopathy. ESC Heart Fail 2025; 12:1256-1270. [PMID: 39499136 PMCID: PMC11911622 DOI: 10.1002/ehf2.15135] [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: 05/21/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND AND AIMS The heart is a metabolic organ rich in mitochondria. The failing heart reprograms to utilize different energy substrates, which increase its oxygen consumption. These adaptive changes contribute to increased oxidative stress. Hypertrophic cardiomyopathy (HCM) is a common heart condition, affecting approximately 15% of the general cat population. Feline HCM shares phenotypical and genotypical similarities with human HCM, but the disease mechanisms for both species are incompletely understood. Our goal was to characterize global changes in metabolome between healthy control cats and cats with different stages of HCM. METHODS Serum samples from 83 cats, the majority (70/83) of which were domestic shorthair and included 23 healthy control cats, 31 and 12 preclinical cats with American College of Veterinary Internal Medicine (ACVIM) stages B1 and B2, respectively, and 17 cats with history of clinical heart failure or arterial thromboembolism (ACVIM stage C), were collected for untargeted metabolomic analysis. Multiple linear regression adjusted for age, sex and body weight was applied to compare between control and across HCM groups. RESULTS Our study identified 1253 metabolites, of which 983 metabolites had known identities. Statistical analysis identified 167 metabolites that were significantly different among groups (adjusted P < 0.1). About half of the differentially identified metabolites were lipids, including glycerophospholipids, sphingolipids and cholesterol. Serum concentrations of free fatty acids, 3-hydroxy fatty acids and acylcarnitines were increased in HCM groups compared with control group. The levels of creatine phosphate and multiple Krebs cycle intermediates, including succinate, aconitate and α-ketoglutarate, also accumulated in the circulation of HCM cats. In addition, serum levels of nicotinamide and tryptophan, precursors for de novo NAD+ biosynthesis, were reduced in HCM groups versus control group. Glutathione metabolism was altered. Serum levels of cystine, the oxidized form of cysteine and cysteine-glutathione disulfide, were elevated in the HCM groups, indicative of heightened oxidative stress. Further, the level of ophthalmate, an endogenous glutathione analog and competitive inhibitor, was increased by more than twofold in HCM groups versus control group. Finally, several uremic toxins, including guanidino compounds and protein bound putrescine, accumulated in the circulation of HCM cats. CONCLUSIONS Our study provided evidence of deranged energy metabolism, altered glutathione homeostasis and impaired renal uremic toxin excretion. Altered lipid metabolism suggested perturbed structure and function of cardiac sarcolemma membrane and lipid signalling.
Collapse
Affiliation(s)
| | - Max Homilius
- Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Erin Achilles
- Department of Clinical Sciences and Advanced MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura K. Massey
- Department of Clinical Sciences and Advanced MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Victoria Convey
- Department of Clinical Sciences and Advanced MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Åsa Ohlsson
- Department of Animal BiosciencesSwedish University of Agricultural SciencesUppsalaSweden
| | - Ingrid Ljungvall
- Department of Clinical SciencesSwedish University of Agricultural SciencesUppsalaSweden
| | - Jens Häggström
- Department of Clinical SciencesSwedish University of Agricultural SciencesUppsalaSweden
| | | | | | - Sharlene Day
- Division of Cardiovascular Medicine, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Calum A. MacRae
- Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Mark A. Oyama
- Department of Clinical Sciences and Advanced MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Gaia de Sousa F, Mendes ACR, de Carvalho LP, Beier SL. Clinical-Diagnostic and Therapeutic Advances in Feline Hypertrophic Cardiomyopathy. Vet Sci 2025; 12:289. [PMID: 40267000 PMCID: PMC11946439 DOI: 10.3390/vetsci12030289] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
A comprehensive literature review was conducted to summarise existing evidence and the latest therapeutic advancements in feline HCM. The disease phenotype is characterised by the development of concentric hypertrophy, which is variable but often asymmetric, primarily affecting the left ventricle. The HCM is marked by diastolic dysfunction, resulting in a reduced intracavitary internal diameter due to the disordered alignment of cardiomyocytes and excessive collagen deposition, which thickens the myocardial tissue and impairs relaxation. There is strong evidence supporting the role of genetic mutations in sarcomeric genes in the development of HCM. Clinical signs vary but are frequently associated with congestive heart failure (CHF) and ATE. Diagnosis is based on a combination of clinical evaluation, laboratory tests, and imaging studies, although certain aspects of the disease remain insufficiently understood and require further research. Therapeutic management focuses on controlling clinical signs, slowing disease progression, and improving both quality of life and life expectancy. However, ongoing studies are essential to refine diagnostic strategies and explore novel treatment options for better disease management.
Collapse
Affiliation(s)
- Felipe Gaia de Sousa
- Department of Veterinary Clinic and Surgery, Veterinary School, Federal University of Minas Gerais—UFMG, Belo Horizonte 31620-295, Minas Gerais, Brazil;
| | - Ana Cristina Ribeiro Mendes
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Pontifical Catholic University of Minas Gerais—PUC Minas, Belo Horizonte 30140-002, Minas Gerais, Brazil; (A.C.R.M.); (L.P.d.C.)
| | - Luisa Pimenta de Carvalho
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Pontifical Catholic University of Minas Gerais—PUC Minas, Belo Horizonte 30140-002, Minas Gerais, Brazil; (A.C.R.M.); (L.P.d.C.)
| | - Suzane Lilian Beier
- Department of Veterinary Clinic and Surgery, Veterinary School, Federal University of Minas Gerais—UFMG, Belo Horizonte 31620-295, Minas Gerais, Brazil;
| |
Collapse
|
3
|
Oleynikov D. Myocardial expression of connexin 43 in cats with hypertrophic and restrictive cardiomyopathy phenotype. Open Vet J 2025; 15:1244-1252. [PMID: 40276190 PMCID: PMC12017731 DOI: 10.5455/ovj.2025.v15.i3.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/20/2025] [Indexed: 04/26/2025] Open
Abstract
Background In humans and cats hypertrophic cardiomyopathy (HCM) is a cause of sudden cardiac death. This is usually associated with arrhythmias, based on myocardial fibrosis and electric impulse propagation disturbances. Restrictive cardiomyopathy (RCM) is a CM associated with excessive fibrosis which is predisposed to arrhythmic episodes. Electric coupling in the myocardium is based on the His-Purkinje system and cardiomyocytes cell-to-cell contacts. Cell connection is based on gap junctions and their structural proteins-connexins. Today there is a lack of information in the literature regarding these functional units and their distribution in cats. Aim Discover the presence of connexin43 (Cx43) in myocardial tissues of cats and to differentiate in HCM and RCM phenotypes. Methods Retrospective analysis of materials collected from cats with CM. Results Animals with the histological animals with histological and immunohistochemical markers of HCM and RCM. Cx43 was distributed in the myocardial tissue of a healthy cat in a typical pattern to other described animals (rats, mice, and human). In HCM, Cx43 was decreased and lateralized near the fibrotic zones, and it was absent in the scar tissue. In RCM, there was a similar pattern, but loci with spontaneously altered expression of Cx43 were also observed, forming lacunas in the gap junction or presented as an intermittent granulated mass. Conclusion Cx43 has different expression patterns in different CM phenotypes; however, the role of this fact in arrhythmogenesis needs to be studied.
Collapse
|
4
|
Janus-Ziółkowska I, Bubak J, Ciaputa R, Kandefer-Gola M, Noszczyk-Nowak A. The Pathological and Histopathological Findings in Cats with Clinically Recognised Hypertrophic Cardiomyopathy Are Related to the Severity of Clinical Signs and Disease Duration. Animals (Basel) 2025; 15:703. [PMID: 40075986 PMCID: PMC11898935 DOI: 10.3390/ani15050703] [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: 01/28/2025] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most frequent type of cardiac disease in cats. Due to its high prevalence and risk of sudden and severe signs, the disease is an important topic of various research. Despite the focus on the clinical course of the disease, studies presenting the pathological and histopathological patterns are rare. The study was conducted as a retrospective analysis of feline patients subjected to postmortem examination in the Cardiopathology Unit due to a clinical diagnosis of hypertrophic cardiomyopathy based on echocardiographic examination and ACVIM guidelines. Thirty-four cats clinically diagnosed with HCM were enrolled in the study. During the postmortem examination, hearts were subjected to gross morphometric and histopathological evaluation. Our results show that the histopathological pattern in cats with clinically stated HCM is very diverse, affecting both ventricles and atria. The histopathological picture is more complex in animals diagnosed earlier and treated for a longer period. Moreover, it is generally unrelated to wall thickness, with only left ventricular fibrosis affecting the thickness of the left ventricular wall. In conclusion, further research combining clinical and pathological results is required to unambiguously determine the histopathological remodelling that takes place in the myocardium of cats with hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Izabela Janus-Ziółkowska
- Department of Pathology, Wrocław University of Environmental and Life Sciences, CK Norwida 31, 50-375 Wrocław, Poland; (J.B.); (R.C.); (M.K.-G.)
| | - Joanna Bubak
- Department of Pathology, Wrocław University of Environmental and Life Sciences, CK Norwida 31, 50-375 Wrocław, Poland; (J.B.); (R.C.); (M.K.-G.)
| | - Rafał Ciaputa
- Department of Pathology, Wrocław University of Environmental and Life Sciences, CK Norwida 31, 50-375 Wrocław, Poland; (J.B.); (R.C.); (M.K.-G.)
| | - Małgorzata Kandefer-Gola
- Department of Pathology, Wrocław University of Environmental and Life Sciences, CK Norwida 31, 50-375 Wrocław, Poland; (J.B.); (R.C.); (M.K.-G.)
| | - Agnieszka Noszczyk-Nowak
- Department of Internal Medicine and Clinic of Diseases of Horses, Dogs and Cats, Wrocław University of Environmental and Life Sciences, Grunwaldzki Sq 47, 50-366 Wrocław, Poland
| |
Collapse
|
5
|
Matsumoto Y, Yik-Lok Chung C, Isobe S, Sakamoto M, Lin X, Chan TF, Hirakawa H, Ishihara G, Lam HM, Nakayama S, Sasamoto S, Tanizawa Y, Watanabe A, Watanabe K, Yagura M, Niimura Y, Nakamura Y. Chromosome-scale assembly with improved annotation provides insights into breed-wide genomic structure and diversity in domestic cats. J Adv Res 2024:S2090-1232(24)00478-8. [PMID: 39490737 DOI: 10.1016/j.jare.2024.10.023] [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: 02/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION Comprehensive genomic resources offer insights into biological features, including traits/disease-related genetic loci. The current reference genome assembly for the domestic cat (Felis catus), Felis_Catus_9.0 (felCat9), derived from sequences of the Abyssinian cat, may inadequately represent the general cat population, limiting the extent of deducible genetic variations. OBJECTIVES The goal was to develop Anicom American Shorthair 1.0 (AnAms1.0), a reference-grade chromosome-scale cat genome assembly. METHODS In contrast to prior assemblies relying on Abyssinian cat sequences, AnAms1.0 was constructed from the sequences of more popular American Shorthair breed, which is related to more breeds than the Abyssinian cat. By combining advanced genomics technologies, including PacBio long-read sequencing and Hi-C- and optical mapping data-based sequence scaffolding, we compared AnAms1.0 to existing Felidae genome assemblies (20 scaffolds, scaffolds N50 > 150 Mbp). Homology-based and ab initio gene annotation through Iso-Seq and RNA-Seq was used to identify new coding genes and splice variants. RESULTS AnAms1.0 demonstrated superior contiguity and accuracy than existing Felidae genome assemblies. Using AnAms1.0, we identified over 1.5 thousand structural variants and 29 million repetitions compared to felCat9. Additionally, we identified > 1,600 novel protein-coding genes. Notably, olfactory receptor structural variants and cardiomyopathy-related variants were identified. CONCLUSION AnAms1.0 facilitates the discovery of novel genes related to normal and disease phenotypes in domestic cats. The analyzed data are publicly accessible on Cats-I (https://cat.annotation.jp/), which we established as a platform for accumulating and sharing genomic resources to discover novel genetic traits and advance veterinary medicine.
Collapse
Affiliation(s)
- Yuki Matsumoto
- Research and Development Section, Anicom Specialty Medical Institute Inc., Yokohama, Kanagawa, Japan; Data Science Center, Azabu University, Sagamihara, Kanagawa, Japan.
| | - Claire Yik-Lok Chung
- School of Life Sciences and the Center for Soybean Research of the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Sachiko Isobe
- Kazusa DNA Research Institute, Kisarazu, Chiba, Japan
| | - Mika Sakamoto
- National Institute of Genetics, Research Organization of Information and Systems, Mishima, Shizuoka, Japan
| | - Xiao Lin
- School of Life Sciences and the Center for Soybean Research of the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Ting-Fung Chan
- School of Life Sciences and the Center for Soybean Research of the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | | | - Genki Ishihara
- Research and Development Section, Anicom Specialty Medical Institute Inc., Yokohama, Kanagawa, Japan
| | - Hon-Ming Lam
- School of Life Sciences and the Center for Soybean Research of the State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | | | | | - Yasuhiro Tanizawa
- National Institute of Genetics, Research Organization of Information and Systems, Mishima, Shizuoka, Japan
| | | | - Kei Watanabe
- Research and Development Section, Anicom Specialty Medical Institute Inc., Yokohama, Kanagawa, Japan
| | - Masaru Yagura
- National Institute of Genetics, Research Organization of Information and Systems, Mishima, Shizuoka, Japan
| | - Yoshihito Niimura
- Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Yasukazu Nakamura
- National Institute of Genetics, Research Organization of Information and Systems, Mishima, Shizuoka, Japan.
| |
Collapse
|
6
|
Guelfi G, Venanzi N, Capaccia C, Stefanetti V, Brachelente C, Sforna M, Porciello F, Lepri E. Feline hypertrophic cardiomyopathy: Does the microRNA-mRNA regulatory network contribute to heart sarcomeric protein remodelling? Int J Exp Pathol 2024; 105:170-183. [PMID: 39138588 PMCID: PMC11574647 DOI: 10.1111/iep.12514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
Feline primary hypertrophic cardiomyopathy (HCM) is an intrinsic myocardial disease characterized by concentric hypertrophy of the left ventricle. In the present study, we investigated the microRNA-mRNA regulatory network in feline myocardial tissue affected by primary (HCMI) and secondary HCM (HCMII). MRNA expression levels of sarcomeric genes, including, TNNT2, TNNI3, MYH7, MYBPC3, TPM1 and ACTC1 were assessed in the FFPE myocardial tissues. FFPE tissues from healthy cats were sequenced by the NGS, to explore, in the entire non-deposited miRNome, the expression level of microRNAs targeting the complementary sequences of selected sarcomeric mRNAs. The sarcomeric genes TNNT2, MYH7, MYBPC3 and TPM1 showed a statistically significant upregulation in HCMI compared to HCMII (p < .01), except ACTC1 which was downregulated (p < .01); TNNI3 showed no statistically significant difference. In HCMII miR-122-5p, miR-338-3p, miR-484, miR-370-3p, miR-92b-3p, miR-375 and miR-370-3p showed a significant upregulation (p < .01) compared to control. The exception was miR-30a-5p which showed downregulation. Worthy of note is the 4-fold higher expression of miR-370-3p, a key regulator of MYBPC3, in HMCI compared to HMCII. This research does not solve the aetiological mystery of HCM, but it may help to find a way to help diagnose and define the prognosis of HCM in cats.
Collapse
Affiliation(s)
- Gabriella Guelfi
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Noemi Venanzi
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Camilla Capaccia
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Valentina Stefanetti
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Chiara Brachelente
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Monica Sforna
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Francesco Porciello
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| | - Elvio Lepri
- Department of Veterinary Medicine, Università Degli Studi di Perugia, Perugia, Italy
| |
Collapse
|
7
|
Grzeczka A, Graczyk S, Pasławski R, Pasławska U. Genetic Basis of Hypertrophic Cardiomyopathy in Cats. Curr Issues Mol Biol 2024; 46:8752-8766. [PMID: 39194734 DOI: 10.3390/cimb46080517] [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: 07/01/2024] [Revised: 07/21/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common cardiovascular condition in cats, affecting yth males and females of all ages. Some breeds, such as Ragdolls and Maine Coons, can develop HCM at a young age. The disease has a wide range of progression and severity, characterized by various pathological changes in the heart, including arteritis, fibrous tissue deposition, and myocardial cell hypertrophy. Left ventricular hypertrophy, which can restrict blood flow, is a common feature of HCM. The disease may persist into old age and eventually lead to heart failure and increased diastolic pressure. The basis of HCM in cats is thought to be genetic, although the exact mechanisms are not fully understood. Mutations in sarcomeric proteins, in particular myosin-binding protein C (MYBPC3), have been identified in cats with HCM. Two specific mutations, MYBPC3 [R818W] and MYBPC3 [A31P], have been classified as 'pathogenic'. Other variants in genes such as MYBPC3, TNNT2, ALMS1, and MYH7 are also associated with HCM. However, there are cases where cats without known genetic mutations still develop HCM, suggesting the presence of unknown genetic factors contributing to the disease. This work aims to summarise the new knowledge of HCM in cats and the alterations in cardiac tissue as a result of genetic variants.
Collapse
Affiliation(s)
- Arkadiusz Grzeczka
- Department for Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Szymon Graczyk
- Department for Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Robert Pasławski
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Urszula Pasławska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| |
Collapse
|
8
|
Pearce A, Ponnam S, Holt MR, Randall T, Beckingham R, Kho AL, Kampourakis T, Ehler E. Missense mutations in the central domains of cardiac myosin binding protein-C and their potential contribution to hypertrophic cardiomyopathy. J Biol Chem 2024; 300:105511. [PMID: 38042491 PMCID: PMC10772716 DOI: 10.1016/j.jbc.2023.105511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/05/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023] Open
Abstract
Myosin binding protein-C (MyBP-C) is a multidomain protein that regulates muscle contraction. Mutations in MYBPC3, the gene encoding for the cardiac variant (henceforth called cMyBP-C), are amongst the most frequent causes of hypertrophic cardiomyopathy. Most mutations lead to a truncated version of cMyBP-C, which is most likely unstable. However, missense mutations have also been reported, which tend to cluster in the central domains of the cMyBP-C molecule. This suggests that these central domains are more than just a passive spacer between the better characterized N- and C-terminal domains. Here, we investigated the potential impact of four different missense mutations, E542Q, G596R, N755K, and R820Q, which are spread over the domains C3 to C6, on the function of MyBP-C on both the isolated protein level and in cardiomyocytes in vitro. Effect on domain stability, interaction with thin filaments, binding to myosin, and subcellular localization behavior were assessed. Our studies show that these missense mutations result in slightly different phenotypes at the molecular level, which are mutation specific. The expected functional readout of each mutation provides a valid explanation for why cMyBP-C fails to work as a brake in the regulation of muscle contraction, which eventually results in a hypertrophic cardiomyopathy phenotype. We conclude that missense mutations in cMyBP-C must be evaluated in context of their domain localization, their effect on interaction with thin filaments and myosin, and their effect on protein stability to explain how they lead to disease.
Collapse
Affiliation(s)
- Amy Pearce
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom; British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Saraswathi Ponnam
- British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics (School of Basic and Biosciences), King's College London, London, United Kingdom
| | - Mark R Holt
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom; British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Thomas Randall
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom; British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Rylan Beckingham
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom; British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom
| | - Ay Lin Kho
- British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics (School of Basic and Biosciences), King's College London, London, United Kingdom
| | - Thomas Kampourakis
- British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics (School of Basic and Biosciences), King's College London, London, United Kingdom
| | - Elisabeth Ehler
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, United Kingdom; British Heart Foundation Centre of Research Excellence, King's College London, London, United Kingdom; Randall Centre for Cell and Molecular Biophysics (School of Basic and Biosciences), King's College London, London, United Kingdom.
| |
Collapse
|
9
|
Lo ST, Li RHL, Georges CJ, Nguyen N, Chen CK, Stuhlmann C, Oldach MS, Rivas VN, Fousse S, Harris SP, Stern JA. Synergistic inhibitory effects of clopidogrel and rivaroxaban on platelet function and platelet-dependent thrombin generation in cats. J Vet Intern Med 2023; 37:1390-1400. [PMID: 37208839 PMCID: PMC10365033 DOI: 10.1111/jvim.16727] [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] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/07/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Dual antithrombotic treatment (DAT) with clopidogrel and rivaroxaban sometimes is prescribed to cats with hypertrophic cardiomyopathy at risk of thromboembolism. To date, no studies have evaluated their combined effects on platelet function. OBJECTIVES/HYPOTHESIS Evaluate the safety of DAT in healthy cats and compare, ex vivo, platelet-dependent thrombin generation and agonist-induced platelet activation and aggregation in cats treated with clopidogrel, rivaroxaban, or DAT. We hypothesized that DAT would safely modulate agonist-induced platelet activation and aggregation more effectively than single agent treatment. ANIMALS Nine apparently healthy 1-year-old cats selected from a research colony. METHODS Unblinded, nonrandomized ex vivo cross-over study. All cats received 7 days of rivaroxaban (0.6 ± 0.1 mg/kg PO), clopidogrel (4.7 ± 0.8 mg/kg PO), or DAT with defined washout periods between treatments. Before and after each treatment, adenosine diphosphate (ADP)- and thrombin-induced platelet P-selectin expression was evaluated using flow cytometry to assess platelet activation. Platelet-dependent thrombin generation was measured by fluorescence assay. Platelet aggregation was assessed using whole blood impedance platelet aggregometry. RESULTS No cats exhibited adverse effects. Of the 3 treatments, only DAT significantly decreased the number of activated platelets (P = .002), modulated platelet activation in response to thrombin (P = .01), dampened thrombin generation potential (P = .01), and delayed maximum reaction velocity (P = .004) in thrombin generation. Like clopidogrel, DAT inhibited ADP-mediated platelet aggregation. However, rivaroxaban alone resulted in increased aggregation and activation in response to ADP. CONCLUSION AND CLINICAL IMPORTANCE Treatment combining clopidogrel and rivaroxaban (DAT) safely decreases platelet activation, platelet response to agonists, and thrombin generation in feline platelets more effectively than monotherapy with either clopidogrel or rivaroxaban.
Collapse
Affiliation(s)
- Sara T. Lo
- University of California Davis School of Veterinary MedicineWilliam R. Prichard Veterinary Medical Teaching HospitalDavisCaliforniaUSA
| | - Ronald H. L. Li
- Surgical and Radiological SciencesUniversity of California, DavisDavisCaliforniaUSA
| | - Catherine J. Georges
- University of California Davis School of Veterinary MedicineWilliam R. Prichard Veterinary Medical Teaching HospitalDavisCaliforniaUSA
| | - Nghi Nguyen
- Surgical and Radiological SciencesUniversity of California Davis School of Veterinary MedicineDavisCaliforniaUSA
| | - Cheyenne K. Chen
- Surgical and Radiological SciencesUniversity of California Davis School of Veterinary MedicineDavisCaliforniaUSA
| | - Claire Stuhlmann
- University of California Davis School of Veterinary MedicineWilliam R. Prichard Veterinary Medical Teaching HospitalDavisCaliforniaUSA
| | | | - Victor Noel Rivas
- Medicine and EpidemiologyUniversity of California Davis School of Veterinary MedicineDavisCaliforniaUSA
| | - Samantha Fousse
- University of California Davis School of Veterinary Medicine – VME, UC Davis 2108 Tupper Hall, One Shields AvenueDavis, California 95616‐5270USA
| | - Samantha P. Harris
- Cellular and Molecular Medicine, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Joshua A. Stern
- Department of Medicine & EpidemiologyUniversity of California, Davis, 2108 Tupper Hall, One Shields AvenueDavis, California 95616USA
| |
Collapse
|
10
|
Cheng WC, Lawson C, Liu HH, Wilkie L, Dobromylskyj M, Luis Fuentes V, Dudhia J, Connolly DJ. Exploration of Mediators Associated with Myocardial Remodelling in Feline Hypertrophic Cardiomyopathy. Animals (Basel) 2023; 13:2112. [PMID: 37443910 DOI: 10.3390/ani13132112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) affects both humans and cats and exhibits considerable interspecies similarities that are exemplified by underlying pathological processes and clinical presentation to the extent that developments in the human field may have direct relevance to the feline disease. Characteristic changes on histological examination include cardiomyocyte hypertrophy and interstitial and replacement fibrosis. Clinically, HCM is characterised by significant diastolic dysfunction due to a reduction in ventricular compliance and relaxation associated with extracellular matrix (ECM) remodelling and the development of ventricular hypertrophy. Studies in rodent models and human HCM patients have identified key protein mediators implicated in these pathological changes, including lumican, lysyl oxidase and TGF-β isoforms. We therefore sought to quantify and describe the cellular location of these mediators in the left ventricular myocardium of cats with HCM and investigate their relationship with the quantity and structural composition of the ECM. We identified increased myocardial content of lumican, LOX and TGF-β2 mainly attributed to their increased expression within cardiomyocytes in HCM cats compared to control cats. Furthermore, we found strong correlations between the expressions of these mediators that is compatible with their role as important components of cellular pathways promoting remodelling of the left ventricular myocardium. Fibrosis and hypertrophy are important pathological changes in feline HCM, and a greater understanding of the mechanisms driving this pathology may facilitate the identification of potential therapies.
Collapse
Affiliation(s)
- Wan-Ching Cheng
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield AL9 7TA, UK
| | - Charlotte Lawson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Hui-Hsuan Liu
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Lois Wilkie
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield AL9 7TA, UK
| | | | - Virginia Luis Fuentes
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield AL9 7TA, UK
| | - Jayesh Dudhia
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield AL9 7TA, UK
| | - David J Connolly
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield AL9 7TA, UK
| |
Collapse
|
11
|
Stern JA, Rivas VN, Kaplan JL, Ueda Y, Oldach MS, Ontiveros ES, Kooiker KB, van Dijk SJ, Harris SP. Hypertrophic cardiomyopathy in purpose-bred cats with the A31P mutation in cardiac myosin binding protein-C. Sci Rep 2023; 13:10319. [PMID: 37365215 PMCID: PMC10293195 DOI: 10.1038/s41598-023-36932-5] [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] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
We sought to establish a large animal model of inherited hypertrophic cardiomyopathy (HCM) with sufficient disease severity and early penetrance for identification of novel therapeutic strategies. HCM is the most common inherited cardiac disorder affecting 1 in 250-500 people, yet few therapies for its treatment or prevention are available. A research colony of purpose-bred cats carrying the A31P mutation in MYBPC3 was founded using sperm from a single heterozygous male cat. Cardiac function in four generations was assessed by periodic echocardiography and measurement of blood biomarkers. Results showed that HCM penetrance was age-dependent, and that penetrance occurred earlier and was more severe in successive generations, especially in homozygotes. Homozygosity was also associated with progression from preclinical to clinical disease. A31P homozygous cats represent a heritable model of HCM with early disease penetrance and a severe phenotype necessary for interventional studies aimed at altering disease progression. The occurrence of a more severe phenotype in later generations of cats, and the occasional occurrence of HCM in wildtype cats suggests the presence of at least one gene modifier or a second causal variant in this research colony that exacerbates the HCM phenotype when inherited in combination with the A31P mutation.
Collapse
Affiliation(s)
- Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1038 William Moore Dr, Raleigh, NC, 27606, USA
| | - Victor N Rivas
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1038 William Moore Dr, Raleigh, NC, 27606, USA
| | - Joanna L Kaplan
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Yu Ueda
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, 1038 William Moore Dr, Raleigh, NC, 27606, USA
| | - Maureen S Oldach
- VCA Sacramento Veterinary Referral Center, 9801 Old Winery Place, Sacramento, CA, 95827, USA
| | - Eric S Ontiveros
- Rady Children's Institute for Genomic Medicine, 7910 Frost Street, San Diego, CA, 92123, USA
| | - Kristina B Kooiker
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, 98109, USA
| | - Sabine J van Dijk
- Department of Neurobiology, Physiology, and Behavior, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Samantha P Harris
- Department of Neurobiology, Physiology, and Behavior, University of California-Davis, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Physiology, College of Medicine-Tucson, University of Arizona, 313 Medical Research Building, 1656 E Mabel St., Tucson, AZ, 85724, USA.
| |
Collapse
|
12
|
Joshua J, Caswell JL, Monné Rodriguez JM, Kipar A, O'Sullivan ML, Wood G, Fonfara S. MicroRNA profiling of the feline left heart identifies chamber-specific expression signatures in health and in advanced hypertrophic cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100037. [PMID: 39801693 PMCID: PMC11708362 DOI: 10.1016/j.jmccpl.2023.100037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common heart disease in humans and cats, nonetheless, the disease pathogenesis is still poorly understood. MicroRNAs are suspected to be involved in the disease process but the myocardial microRNA expression pattern in cats has not been identified. We hypothesized that microRNA profiles differ between healthy cats and cats with HCM. Small RNA sequencing on left ventricle (LV) and left atria (LA) samples from healthy cats (8 LV, 8 LA) and cats with HCM (7 LV, 5 LA) was performed. We identified 1039 differentially expressed microRNAs (False Discovery Rate <0.01, fold change >2). Cats with HCM were found to have a distinct microRNA expression profile with apparent regional heterogeneity. Comparing the HCM and control hearts, we detected 80 differentially expressed microRNAs for the HCM LV, and 37 for the LA. These included LV and LA enriched miR-21, miR-146b, and reduced miR-122-5p, which were recently suggested as key microRNAs for the HCM pathogenesis, and miR-132, which might be of therapeutic interest. Several top enriched microRNAs: miR-3958, miR-382-5p, miR-487a-5p (HCM LV); miR-chrD4_30107-3p (HCM LA); miR-3548 (HCM LV and LA) have either not been reported in the heart or only little is known. We identified potentially relevant microRNAs and further investigations into their role are required. Genes known to be targeted by the differentially expressed microRNAs were associated with inflammation and growth pathways in the HCM LV and LA, cardioprotective pathways in the LV, and fibrosis and structural changes in the LA when compared to healthy hearts.
Collapse
Affiliation(s)
- Jessica Joshua
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph N1G 2W1, Ontario, Canada
| | - Jeff L. Caswell
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
| | - Josep M. Monné Rodriguez
- University of Zurich, Vetsuisse Faculty, Institute of Veterinary Pathology, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Anja Kipar
- University of Zurich, Vetsuisse Faculty, Institute of Veterinary Pathology, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - M. Lynne O'Sullivan
- University of Prince Edward Island, Department of Companion Animals, Charlottetown C1A 4P3, Prince Edward Island, Canada
| | - Geoffrey Wood
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
| | - Sonja Fonfara
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph N1G 2W1, Ontario, Canada
| |
Collapse
|
13
|
Penning LC, van den Boom R. Companion animal organoid technology to advance veterinary regenerative medicine. Front Vet Sci 2023; 10:1032835. [PMID: 37008367 PMCID: PMC10063859 DOI: 10.3389/fvets.2023.1032835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
First year medical and veterinary students are made very aware that drugs can have very different effects in various species or even in breeds of one specific species. On the other hand, the “One Medicine” concept implies that therapeutic and technical approaches are exchangeable between man and animals. These opposing views on the (dis)similarities between human and veterinary medicine are magnified in regenerative medicine. Regenerative medicine promises to stimulate the body's own regenerative capacity via activation of stem cells and/or the application of instructive biomaterials. Although the potential is enormous, so are the hurdles that need to be overcome before large scale clinical implementation is realistic. It is in the advancement of regenerative medicine that veterinary regenerative medicine can play an instrumental and crucial role. This review describes the discovery of (adult) stem cells in domesticated animals, mainly cats and dogs. The promise of cell-mediated regenerative veterinary medicine is compared to the actual achievements, and this will lead to a set of unanswered questions (controversies, research gaps, potential developments in relation to fundamental, pre-clinical, and clinical research). For veterinary regenerative medicine to have impact, either for human medicine and/or for domesticated animals, answering these questions is pivotal.
Collapse
|
14
|
Bastos RF, Tuleski GLR, Sousa MG. QT interval instability and QRS interval dispersion in healthy cats and cats with a hypertrophic cardiomyopathy phenotype. J Feline Med Surg 2023; 25:1098612X231151479. [PMID: 36745542 PMCID: PMC10812083 DOI: 10.1177/1098612x231151479] [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] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Hypertrophic cardiomyopathy (HCM) is the most common heart disease in cats. Electrocardiographic (ECG) analysis can help with the diagnosis of HCM and also in the investigation of the secondary consequences of the disease. This study investigated ECG markers of QT interval variability (total instability [TI], short-term instability [STI], long-term instability [LTI], QT variance [QTv]), mean QT interval (QTa) and QT interval corrected for heart rate (QTac), as well as the duration (QRSd) and dispersion (QRSv) of the QRS interval in healthy cats and in those with HCM. METHODS Data were collected from 63 domestic cats: 40 in the control group and 23 in the HCM group. Fifty consecutive QT intervals were recorded for all cats and then QTa, QTac, QTv, TI, LTI and STI were calculated. QRSd and QRSv were also obtained for all animals. A Mann-Whitney U-test was used for group comparison. Receiver operating characteristic curves were plotted to evaluate the sensitivity and specificity of all markers for HCM. Logistic regression analysis was performed to assess the risks of cats having HCM, based on the studied indexes. RESULTS QTa (P <0.01), QTac (P <0.01), QRSd (P <0.01) and STI (P = 0.02) were higher in the HCM group. QTa >158.8 ms, QTac >27.4 ms and QRSd >0.045 s had an accuracy of 77.4%, 68.2% and 80.9%, respectively, in detecting HCM. Logistic regression showed that cats with QTa >158 ms, QTac >27.4 ms and QRSd >0.045 s had a 1.58-, 1,23- and 6.5-fold higher risk, respectively, of developing HCM. CONCLUSIONS AND RELEVANCE Cats with HCM had higher ventricular instability as assessed by STI and showed a prolongation of the QT and QRS intervals via the QTa, QTac and QRSd markers. These markers show potential as ancillary screening tools for identifying the presence of HCM.
Collapse
Affiliation(s)
- Rodrigo Franco Bastos
- Laboratory of Comparative Cardiology, Department of Veterinary Medicine, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Giovana LR Tuleski
- Laboratory of Comparative Cardiology, Department of Veterinary Medicine, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Marlos Gonçalves Sousa
- Laboratory of Comparative Cardiology, Department of Veterinary Medicine, Federal University of Paraná (UFPR), Curitiba, Brazil
| |
Collapse
|
15
|
Sharpe AN, Oldach MS, Rivas VN, Kaplan JL, Walker AL, Kovacs SL, Hwee DT, Cremin P, Morgan BP, Malik FI, Harris SP, Stern JA. Effects of Aficamten on cardiac contractility in a feline translational model of hypertrophic cardiomyopathy. Sci Rep 2023; 13:32. [PMID: 36593243 PMCID: PMC9807554 DOI: 10.1038/s41598-022-26630-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited cardiac disease in humans and cats and lacks efficacious pharmacologic interventions in the preclinical phase of disease. LV outflow tract obstruction (LVOTO) is commonly observed in HCM-affected patients and is a primary driver of heart failure symptoms and reduced quality of life. Novel small-molecule cardiac myosin inhibitors target actin-myosin interactions to alleviate overactive protein interactions. A prospective, randomized, controlled cross-over study was performed to evaluate pharmacodynamic effects of two doses (0.3 and 1 mg/kg) of a next-in-class cardiac myosin inhibitor, aficamten (CK-3773274, CK-274), on cardiac function in cats with the A31P MYBPC3 mutation and oHCM. Dose-dependent reductions in LV systolic function, LVOT pressure gradient, and isovolumetric relaxation times compared to baseline were observed. Promising beneficial effects of reduced systolic function warrant further studies of this next-in-class therapeutic to evaluate the benefit of long-term administration in this patient population.
Collapse
Affiliation(s)
- Ashley N Sharpe
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Maureen S Oldach
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Victor N Rivas
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Joanna L Kaplan
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Ashley L Walker
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Samantha L Kovacs
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Darren T Hwee
- Research and Non-Clinical Development, Cytokinetics, Inc., South San Francisco, CA, USA
| | - Peadar Cremin
- Research and Non-Clinical Development, Cytokinetics, Inc., South San Francisco, CA, USA
| | - Bradley P Morgan
- Research and Non-Clinical Development, Cytokinetics, Inc., South San Francisco, CA, USA
| | - Fady I Malik
- Research and Non-Clinical Development, Cytokinetics, Inc., South San Francisco, CA, USA
| | - Samantha P Harris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
16
|
Sharpe AN, Oldach MS, Kaplan JL, Rivas V, Kovacs SL, Hwee DT, Morgan BP, Malik FI, Harris SP, Stern JA. Pharmacokinetics of a single dose of Aficamten (CK-274) on cardiac contractility in a A31P MYBPC3 hypertrophic cardiomyopathy cat model. J Vet Pharmacol Ther 2023; 46:52-61. [PMID: 36382714 PMCID: PMC10099566 DOI: 10.1111/jvp.13103] [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: 04/23/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent cardiac disease in cats and lacks efficacious preclinical pharmacologic intervention, prompting investigation of novel therapies. Genetic mutations encoding sarcomeric proteins are implicated in the development of HCM and small molecule myosin inhibitors are an emerging class of therapeutics designed to target the interaction of actin and myosin to alleviate the detrimental effects of inappropriate contractile protein interactions. The purpose of this study was to characterize the pharmacodynamic effects of a single oral dose of the novel cardiac myosin inhibitor aficamten (CK-274) on cardiac function in purpose bred cats with naturally occurring A31P MYBPC3 mutation and a clinical diagnosis of HCM with left ventricular outflow tract obstruction (LVOTO). Five purpose bred cats were treated with aficamten (2 mg/kg) or vehicle and echocardiographic evaluations were performed at 0, 6, 24, and 48 h post-dosing. High dose aficamten (2 mg/kg) reduced left ventricular fractional shortening (LVFS%) by increasing the LV systolic internal dimension (LVIDs) and reduced isovolumic relaxation time (IVRT) compared with baseline without significant adverse effects. The marked reduction in systolic function and reduced IVRT coupled with an increased heart rate in treated cats, suggest a lower dose may be optimal. Further studies to determine optimal dosing of aficamten are indicated.
Collapse
Affiliation(s)
- Ashley N. Sharpe
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Maureen S. Oldach
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Joanna L. Kaplan
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Victor Rivas
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Samantha L. Kovacs
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Darren T. Hwee
- Research and Non‐clinical DevelopmentCytokinetics, IncSouth San FranciscoCaliforniaUSA
| | - Bradley P. Morgan
- Research and Non‐clinical DevelopmentCytokinetics, IncSouth San FranciscoCaliforniaUSA
| | - Fady I. Malik
- Research and Non‐clinical DevelopmentCytokinetics, IncSouth San FranciscoCaliforniaUSA
| | - Samantha P. Harris
- Department of Cellular and Molecular MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Joshua A. Stern
- Department of Medicine and Epidemiology, Davis, School of Veterinary MedicineUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
17
|
Joshua J, Caswell J, O’Sullivan ML, Wood G, Fonfara S. Feline myocardial transcriptome in health and in hypertrophic cardiomyopathy-A translational animal model for human disease. PLoS One 2023; 18:e0283244. [PMID: 36928240 PMCID: PMC10019628 DOI: 10.1371/journal.pone.0283244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in cats, characterized by primary left ventricular hypertrophy. Feline HCM closely resembles human HCM and is suggested as translational animal model for the human disease. A genetic cause is established in humans and suspected for cats, but little is known about the gene expression and pathways involved in the pathogenesis of HCM. To investigate the myocardial transcriptome changes in HCM, RNA sequencing was conducted on left ventricle (LV) and left atrium (LA) samples of healthy cats and cats with HCM (each n = 5; 20 samples). Ingenuity Pathway Analysis was used to determine functional pathways, regulators, and networks. Distinct gene expression profiles were identified in the LV and LA of the feline healthy and HCM myocardium. Analysis of differentially expressed mRNAs (>2 fold; FDR < 0.01) found chamber-specific (LV vs. LA) expression in both healthy and HCM groups, with higher transcriptional activity in the LA. Genes that contribute to the distinct structure and function of each chamber in health and HCM were identified in the regional comparison. The gene expression profiles of HCM compared to healthy hearts revealed disease related genes, including THBS4 and KLHL33 (LV), FAM177B and THRSP (LA), the latter 3 have not been reported for the myocardium so far, as the top differently expressed genes in the HCM heart. Differently expressed genes and functional pathways found in the HCM heart are associated with cardiac remodeling and fibrosis, inflammation, microvascular changes, calcium signaling and cardiac metabolism, with some regional differences. RhoGDI-RhoGTPase signaling, integrin and ILK signaling pathways, the LXR/RXR pathway in the LA, and the PPARα/RXRα, HIF1α and CXCR4 pathways in the LV might be of particular importance in the HCM disease process. This study identified region-specific myocardial gene transcription patterns as well as novel genes and pathways associated with HCM.
Collapse
Affiliation(s)
- Jessica Joshua
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
| | - Jeff Caswell
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - M. Lynne O’Sullivan
- University of Prince Edward Island, Department of Companion Animals, Charlottetown, Prince Edward Island, Canada
| | - Geoffrey Wood
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph, Ontario, Canada
| | - Sonja Fonfara
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
18
|
Demeekul K, Sukumolanan P, Panprom C, Thaisakun S, Roytrakul S, Petchdee S. Echocardiography and MALDI-TOF Identification of Myosin-Binding Protein C3 A74T Gene Mutations Involved Healthy and Mutated Bengal Cats. Animals (Basel) 2022; 12:ani12141782. [PMID: 35883329 PMCID: PMC9312240 DOI: 10.3390/ani12141782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to identify the potential peptide candidates and expected proteins associated with MYBPC3-A74T gene mutations in Bengal cats and determine if peptidome profiles differ between healthy controls and cats with MYBPC3-A74T gene mutations. All animals were evaluated using echocardiography. DNA was isolated and followed by the screening test of MYBPC3 gene mutation. The MALDI-TOF mass spectrometry was conducted for analyzing the targeted peptide and protein patterns. The expected protein candidates were searched for within the NCBI database. Our results demonstrated that the MYBPC3-A74T gene mutation was dominant in Bengal cats but not in domestic shorthair cats. Correlations between baseline characteristics and echocardiographic parameters were discovered in Bengal cats. Mass spectrometry profiles of the candidate proteins were suspected to accompany the cat with the MYBPC3-A74T gene mutation, involving integral protein–membrane, organization of nucleus, DNA replication, and ATP-binding protein. Therefore, MYBPC3-A74T gene mutations occur frequently in Bengal cat populations. The high incidence of homozygotes for the mutation supports the causal nature of the MYBPC3-A74T mutation. In addition, peptidomics analysis was established for the first time under this condition to promise a complementary technique for the future clinical diagnosis of the MYBPC3-A74T mutation associated with physiological variables and cardiac morphology in cats.
Collapse
Affiliation(s)
- Kanokwan Demeekul
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand;
| | - Pratch Sukumolanan
- Veterinary Clinical Studies Program, Graduate School, Kasetsart University, Nakorn Pathom 73140, Thailand;
| | - Chattida Panprom
- Kasetsart University Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Kasetsart University, Nakhon Pathom 73140, Thailand;
| | - Siriwan Thaisakun
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani 12120, Thailand; (S.T.); (S.R.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani 12120, Thailand; (S.T.); (S.R.)
| | - Soontaree Petchdee
- Department of Large Animal and Wildlife Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Nakorn Pathom 73140, Thailand
- Correspondence: ; Tel.: +66-34-351-901-3
| |
Collapse
|
19
|
Schipper T, Ohlsson Å, Longeri M, Hayward JJ, Mouttham L, Ferrari P, Smets P, Ljungvall I, Häggström J, Stern JA, Lyons LA, Peelman LJ, Broeckx BJG. The TNNT2:c.95-108G>A variant is common in Maine Coons and shows no association with hypertrophic cardiomyopathy. Anim Genet 2022; 53:526-529. [PMID: 35634705 DOI: 10.1111/age.13223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/05/2022] [Accepted: 05/15/2022] [Indexed: 12/01/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common and potentially fatal heart disease in many cat breeds. An intronic variant in TNNT2, c.95-108G>A, was recently reported as the cause of HCM in the Maine Coon. The aim of this study was to determine this variant's allele frequency in different populations and its possible association with HCM. Based on 160 Maine Coon samples collected in Belgium, Italy, Sweden and the USA, the variant's allele frequency was estimated to be 0.32. Analysis of the 99 Lives feline whole genome sequencing database showed that the TNNT2 variant also occurs in other breeds, as well as mixed-breed cats. Comparison of 31 affected and 58 healthy cats did not reveal significantly increased odds for HCM in homozygotes. Based on the combined evidence and in agreement with the standards and guidelines for the interpretation of sequence variants, this variant is currently classified as a variant of unknown significance and should not be used for breeding decisions regarding HCM.
Collapse
Affiliation(s)
- Tom Schipper
- Department of Veterinary and Biosciences, Ghent University, Merelbeke, Belgium
| | - Åsa Ohlsson
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Maria Longeri
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Jessica J Hayward
- Department of Biomedical Sciences and Cornell Veterinary Biobank, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Lara Mouttham
- Cornell Veterinary Biobank, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Paolo Ferrari
- Osservatorio Veterinario Italiano Cardiopatie, Azzano S. Paolo, Italy
| | - Pascale Smets
- Small Animal Department, Ghent University, Merelbeke, Belgium
| | - Ingrid Ljungvall
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jens Häggström
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Joshua A Stern
- Department of Medicine and Epidemiology, University of California - Davis, Davis, California, USA
| | - Leslie A Lyons
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| | - Luc J Peelman
- Department of Veterinary and Biosciences, Ghent University, Merelbeke, Belgium
| | - Bart J G Broeckx
- Department of Veterinary and Biosciences, Ghent University, Merelbeke, Belgium
| |
Collapse
|
20
|
Walker AL, Ueda Y, Crofton AE, Harris SP, Stern JA. Ambulatory electrocardiography, heart rate variability, and pharmacologic stress testing in cats with subclinical hypertrophic cardiomyopathy. Sci Rep 2022; 12:1963. [PMID: 35121794 PMCID: PMC8817045 DOI: 10.1038/s41598-022-05999-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/21/2022] [Indexed: 11/09/2022] Open
Abstract
The utility of ambulatory electrocardiography (AECG) to evaluate cats with subclinical hypertrophic cardiomyopathy (HCM) for arrhythmias and heart rate variability (HRV) is not well defined but may provide information regarding risk stratification. This prospective study used AECG to evaluate ectopy and HRV in subclinical HCM cats compared to healthy controls and is the first to implement a pharmacologic cardiac stress test. Twenty-three purpose-bred, Maine coon cross cats (16 HCM, 7 control) underwent 48-h of continuous AECG. Terbutaline (0.2-0.3 mg/kg) was administered orally at 24 and 36 h. Heart rate, ectopy frequency and complexity and HRV parameters, including standard deviation of normal R-R intervals (SDNN), were compared pre-terbutaline and post-terbutaline and across phenotype, genotype and sex. Genotype for an HCM-causative mutation was significantly associated with the frequency of supraventricular (P = 0.033) and ventricular (P = 0.026) ectopy across all cats. Seven HCM cats and zero healthy cats had a sinus arrhythmia. Mean heart rate was significantly higher post-terbutaline (p < 0.0001). HCM cats had significantly greater HRV compared to controls (SDNN: p = 0.0006). Male cats had significantly higher HRV (SDNN: p = 0.0001) and lower mean heart rates (p = 0.0001). HRV decreased post-terbutaline (SDNN: p = 0.0008) and changes in HRV observed between sexes were attenuated by terbutaline.
Collapse
Affiliation(s)
- Ashley L Walker
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, Davis, CA, 95616-8732, USA
| | - Yu Ueda
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - Amanda E Crofton
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, Davis, CA, 95616-8732, USA
| | - Samantha P Harris
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Joshua A Stern
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California-Davis, 2108 Tupper Hall, Davis, CA, 95616-8732, USA.
| |
Collapse
|
21
|
Rodríguez JMM, Fonfara S, Hetzel U, Kipar A. Feline hypertrophic cardiomyopathy: reduced microvascular density and involvement of CD34+ interstitial cells. Vet Pathol 2021; 59:269-283. [PMID: 34955067 PMCID: PMC8928422 DOI: 10.1177/03009858211062631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The sequence of pathological events in feline hypertrophic cardiomyopathy (fHCM) is still largely unknown, although we know that fHCM is characterized by interstitial remodeling in a macrophage-driven pro-inflammatory environment and that myocardial ischemia might contribute to its progression. This study aimed to gain further insights into the structural changes associated with interstitial remodeling in fHCM with special focus on the myocardial microvasculature and the phenotype of the interstitial cells. Twenty-eight hearts (16 hearts with fHCM and 12 without cardiac disease) were evaluated in the current study, with immunohistochemistry, RNA-in situ hybridization, and transmission electron microscopy. Morphometrical evaluations revealed a statistically significant lower microvascular density in fHCM. This was associated with structural alterations in capillaries that go along with a widening of the interstitium due to the accumulation of edema fluid, collagen fibers, and mononuclear cells that also proliferated locally. The interstitial cells were mainly of fibroblastic or vascular phenotype, with a substantial contribution of predominantly resident macrophages. A large proportion expressed CD34 mRNA, which suggests a progenitor cell potential. Our results indicate that microvascular alterations are key events in the pathogenesis of fHCM and that myocardial interstitial cell populations with CD34+ phenotype play a role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Josep M Monné Rodríguez
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland.,University of Bern, Bern, Switzerland
| | - Sonja Fonfara
- The Veterinary Cardiac Pathophysiology Consortium.,University of Guelph, Guelph, Ontario, Canada
| | - Udo Hetzel
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland
| | - Anja Kipar
- The Veterinary Cardiac Pathophysiology Consortium.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Investigating Associations Among Relatedness, Genetic Diversity, and Causes of Mortality In Southern Sea Otters (Enhydra lutris nereis). J Wildl Dis 2021; 58:63-75. [PMID: 34818404 DOI: 10.7589/jwd-d-21-00019] [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: 02/05/2021] [Accepted: 07/15/2021] [Indexed: 11/20/2022]
Abstract
Southern sea otter (Enhydra lutris nereis) population recovery is influenced by a variety of factors, including predation, biotoxin exposure, infectious disease, oil spills, habitat degradation, and resource limitation. This population has also experienced a significant genetic bottleneck, resulting in low genetic diversity. We investigated how two metrics, familial relatedness and genetic diversity, are correlated with common causes of mortality in southern sea otters, including cardiomyopathy, acanthocephalan (Profilicollis spp.) peritonitis, systemic protozoal infection (Toxoplasma gondii and Sarcocystis neurona), domoic acid intoxication, end-lactation syndrome, and shark bite. Microsatellite genetic markers were used to examine this association in 356 southern sea otters necropsied from 1998 to 2012. Significant associations with genetic diversity or familial relatedness (P<0.05) were observed for cardiomyopathy, acanthocephalan peritonitis, and sarcocystosis, and these associations varied by sex. Adult male cardiomyopathy cases (n=86) were more related than the null expectation (P<0.049). Conversely, female acanthocephalan peritonitis controls (n=110) were more related than the null expectation (P<0.004). Including genetic diversity as a predictor for fatal acanthocephalan peritonitis in the multivariate logistic model significantly improved model fit; lower genetic diversity was associated with reduced odds of sea otter death due to acanthocephalan peritonitis. Finally, male sarcocystosis controls (n=158) were more related than the null expectation (P<0.011). Including genetic diversity in the multivariate logistic model for fatal S. neurona infection improved model fit; lower genetic diversity was associated with increased odds of sea otter death due to S. neurona. Our study suggests that genetic diversity and familial relatedness, in conjunction with other factors such as age and sex, may influence outcome (survival or death) in relation to several common southern sea otter diseases. Our findings can inform policy for conservation management, such as potential reintroduction efforts, as part of species recovery.
Collapse
|
23
|
Abstract
PRACTICAL RELEVANCE The feline cardiomyopathies are the most prevalent type of heart disease in adult domestic cats. Several forms have been identified (see Parts 2 and 3), with hypertrophic cardiomyopathy (HCM) being the most common. Clinically the cardiomyopathies are often indistinguishable. Cats with subclinical cardiomyopathy may or may not have characteristic physical examination findings (eg, heart murmur, gallop sound), or radiographic cardiomegaly. Cats with severe disease may develop signs of heart failure (eg, dyspnea, tachypnea) or systemic arterial thromboembolism (ATE; eg, pain and paralysis). Sudden death is possible. Treatment usually does not alter the progression from subclinical to clinical disease and often the treatment approach, once clinical signs are apparent, is the same regardless of the type of cardiomyopathy. However, differentiating cardiomyopathy from normal variation may be important prognostically. PATIENT GROUP Domestic cats of any age from 3 months upward, of either sex and of any breed, can be affected. Mixed-breed cats are most commonly affected but certain breeds are disproportionately prone to developing HCM. DIAGNOSTICS Subclinical feline cardiomyopathies may be suspected based on physical examination findings, thoracic radiographs and cardiac biomarker results but often the disease is clinically silent. The definitive clinical confirmatory test is echocardiography. Left heart failure (pulmonary edema and/or pleural effusion) is most commonly diagnosed radiographically, but point-of-care ultrasound and amino terminal pro-B-type natriuretic peptide (NT-proBNP) biomarker testing can also be useful, especially when the stress of taking radiographs is best avoided. KEY FINDINGS Knowledge of pathophysiological mechanisms helps the practitioner identify the feline cardiomyopathies and understand how these diseases progress and how they manifest clinically (heart failure, ATE). Existing diagnostic tests have strengths and limitations, and being aware of these can help a practitioner deliver optimal recommendations regarding referral. CONCLUSIONS Several types of feline cardiomyopathies exist in both subclinical (mild to severe disease) and clinical (severe disease) phases. Heart failure and ATE are the most common clinical manifestations of severe cardiomyopathy and are therapeutic targets regardless of the type of cardiomyopathy. The long-term prognosis is often guarded or poor once overt clinical manifestations are present. AREAS OF UNCERTAINTY Some cats with presumed cardiomyopathy do not have echocardiographic features that fit the classic cardiomyopathies (cardiomyopathy - nonspecific phenotype). Although no definitive treatment is usually available, understanding how cardiomyopathies evolve remains worthy of investigation.
Collapse
Affiliation(s)
- Mark D Kittleson
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, and Veterinary Information Network, 777 West Covell Boulevard, Davis, CA 95616, USA
| | - Etienne Côté
- Department of Companion Animals, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| |
Collapse
|
24
|
Abstract
Practical relevance: Hypertrophic cardiomyopathy (HCM) is the most common form of feline
cardiomyopathy observed clinically and may affect up to approximately 15% of
the domestic cat population, primarily as a subclinical disease.
Fortunately, severe HCM, leading to heart failure or arterial
thromboembolism (ATE), only occurs in a small proportion of these cats. Patient group: Domestic cats of any age from 3 months upward, of either sex and of any
breed, can be affected. A higher prevalence in male and domestic shorthair
cats has been reported. Diagnostics: Subclinical feline HCM may or may not produce a heart murmur or gallop sound.
Substantial left atrial enlargement can often be identified radiographically
in cats with severe HCM. Biomarkers should not be relied on solely to
diagnose the disease. While severe feline HCM can usually be diagnosed via
echocardiography alone, feline HCM with mild to moderate left ventricular
(LV) wall thickening is a diagnosis of exclusion, which means there is no
definitive test for HCM in these cats and so other disorders that can cause
mild to moderate LV wall thickening (eg, hyperthyroidism, systemic
hypertension, acromegaly, dehydration) need to be ruled out. Key findings: While a genetic cause of HCM has been identified in two breeds and is
suspected in another, for most cats the cause is unknown. Systolic anterior
motion of the mitral valve (SAM) is the most common cause of dynamic left
ventricular outflow tract obstruction (DLVOTO) and, in turn, the most common
cause of a heart murmur with feline HCM. While severe DLVOTO is probably
clinically significant and so should be treated, lesser degrees probably are
not. Furthermore, since SAM can likely be induced in most cats with HCM, the
distinction between HCM without obstruction and HCM with obstruction (HOCM)
is of limited importance in cats. Diastolic dysfunction, and its
consequences of abnormally increased atrial pressure leading to signs of
heart failure, and sluggish atrial blood flow leading to ATE, is the primary
abnormality that causes clinical signs and death in affected cats. Treatment
(eg, loop diuretics) is aimed at controlling heart failure. Preventive
treatment (eg, antithrombotic drugs) is aimed at reducing the risk of
complications (eg, ATE). Conclusions: Most cats with HCM show no overt clinical signs and live a normal or
near-normal life despite this disease. However, a substantial minority of
cats develop overt clinical signs referable to heart failure or ATE that
require treatment. For most cats with clinical signs caused by HCM, the
long-term prognosis is poor to grave despite therapy. Areas of uncertainty: Genetic mutations (variants) that cause HCM have been identified in a few
breeds, but, despite valiant efforts, the cause of HCM in the vast majority
of cats remains unknown. No treatment currently exists that reverses or even
slows the cardiomyopathic process in HCM, again despite valiant efforts. The
search goes on.
Collapse
Affiliation(s)
- Mark D Kittleson
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, and Veterinary Information Network, 777 West Covell Boulevard, Davis, CA 95616, USA
| | - Etienne Côté
- Department of Companion Animals, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Prince Edward Island, Canada
| |
Collapse
|
25
|
Bach MBT, Grevsen JR, Kiely MAB, Willesen JL, Koch J. Detection of congestive heart failure by mitral annular displacement in cats with hypertrophic cardiomyopathy - concordance between tissue Doppler imaging-derived tissue tracking and M-mode. J Vet Cardiol 2021; 36:153-168. [PMID: 34298446 DOI: 10.1016/j.jvc.2021.06.003] [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: 10/27/2020] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The left ventricular systolic longitudinal function, traditionally measured by M-mode-derived mitral annular plane systolic excursion (MAPSE), is reduced in feline hypertrophic cardiomyopathy (HCM) and further reduced in cats with left-sided congestive heart failure (CHF). The objectives of this study were to compare longitudinal displacement measured by tissue tracking (TT-LD) and MAPSE in feline HCM and assess these methods' ability to differentiate CHF from preclinical HCM. A further objective was to provide preliminary reference intervals for TT-LD. ANIMALS Eighty-five client-owned cats. METHODS A retrospective case-control study. Anatomical M-mode was used to record MAPSE, and TT-LD was recorded by tissue tracking. RESULTS Reduced longitudinal displacement measured by either MAPSE or TT-LD was significantly associated with CHF in cats with HCM (p < 0.036). Receiver-operating characteristic analysis indicated that TT-LD (AUC: 92.9%-97.9%) was more sensitive and specific than MAPSE (AUC: 85.8%-89.1%) for the detection of CHF. A diagnostic cut-off of 2.89 mm for maximal TT-LD in the left ventricular septum resulted in a sensitivity and specificity of 100% and 83.3%, while a diagnostic cut-off of 2.41 mm in the left ventricular posterior wall resulted in a sensitivity of 100% and a specificity of 90%. CONCLUSIONS M-mode-derived mitral annular plane systolic excursion and TT-LD were strongly correlated, but not interchangeable. Longitudinal displacement measured by tissue tracking decreased more with disease severity than traditional MAPSE. Longitudinal displacement may help detect CHF in cats with HCM - with the maximal TT-LD of the left ventricular posterior wall achieving the highest AUC value.
Collapse
Affiliation(s)
- M B T Bach
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870, Frederiksberg, Denmark.
| | - J R Grevsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870, Frederiksberg, Denmark
| | - M A B Kiely
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870, Frederiksberg, Denmark
| | - J L Willesen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870, Frederiksberg, Denmark
| | - J Koch
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870, Frederiksberg, Denmark
| |
Collapse
|
26
|
Brugada-Terradellas C, Hellemans A, Brugada P, Smets P. Sudden cardiac death: A comparative review of humans, dogs and cats. Vet J 2021; 274:105696. [PMID: 34148018 DOI: 10.1016/j.tvjl.2021.105696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Sudden death is one of the most common causes of death in humans in Western countries. Approximately 85% of these cases are of cardiac origin. In dogs and cats, sudden cardiac death (SCD) also commonly occurs, but fewer pathophysiological and prevalence data are available. Both structural, primarily 'electrical' and ischemic heart diseases are known to cause SCD, many of which share similar underlying arrhythmogenic mechanisms between humans and companion animals. As for underlying genetics, numerous mutations on multiple loci have been related to SCD in humans, but only a few mutations associated with dilated cardiomyopathy and SCD have been identified in dogs, e.g. in the phospholamban and titin genes. Information published from human medicine can therefore inform future veterinary studies, but also dogs and cats could act as spontaneous models of SCD in humans. Further research in both fields is therefore warranted to better understand the pathophysiology, genetics, and prevention of SCD.
Collapse
Affiliation(s)
- Celine Brugada-Terradellas
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | - Arnaut Hellemans
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | - Pedro Brugada
- Pedro Brugada, Cardiovascular Division, UZ Brussel - VUB, Avenue du Laerbeek 101, 1090 Brussels, Belgium
| | - Pascale Smets
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
27
|
A domestic cat whole exome sequencing resource for trait discovery. Sci Rep 2021; 11:7159. [PMID: 33785770 PMCID: PMC8009874 DOI: 10.1038/s41598-021-86200-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Over 94 million domestic cats are susceptible to cancers and other common and rare diseases. Whole exome sequencing (WES) is a proven strategy to study these disease-causing variants. Presented is a 35.7 Mb exome capture design based on the annotated Felis_catus_9.0 genome assembly, covering 201,683 regions of the cat genome. Whole exome sequencing was conducted on 41 cats with known and unknown genetic diseases and traits, of which ten cats had matching whole genome sequence (WGS) data available, used to validate WES performance. At 80 × mean exome depth of coverage, 96.4% of on-target base coverage had a sequencing depth > 20-fold, while over 98% of single nucleotide variants (SNVs) identified by WGS were also identified by WES. Platform-specific SNVs were restricted to sex chromosomes and a small number of olfactory receptor genes. Within the 41 cats, we identified 31 previously known causal variants and discovered new gene candidate variants, including novel missense variance for polycystic kidney disease and atrichia in the Peterbald cat. These results show the utility of WES to identify novel gene candidate alleles for diseases and traits for the first time in a feline model.
Collapse
|
28
|
Meurs KM, Williams BG, DeProspero D, Friedenberg SG, Malarkey DE, Ezzell JA, Keene BW, Adin DB, DeFrancesco TC, Tou S. A deleterious mutation in the ALMS1 gene in a naturally occurring model of hypertrophic cardiomyopathy in the Sphynx cat. Orphanet J Rare Dis 2021. [PMID: 33639992 DOI: 10.1186/s13023-021-01740-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
BACKGROUND Familial hypertrophic cardiomyopathy is a common inherited cardiovascular disorder in people. Many causal mutations have been identified, but about 40% of cases do not have a known causative mutation. Mutations in the ALMS1 gene are associated with the development of Alstrom syndrome, a multisystem familial disease that can include cardiomyopathy (dilated, restrictive). Hypertrophic cardiomyopathy has not been described. The ALMS1 gene is a large gene that encodes for a ubiquitously expressed protein. The function of the protein is not well understood although it is believed to be associated with energy metabolism and homeostasis, cell differentiation and cell cycle control. The ALMS1 protein has also been shown to be involved in the regulation of cell cycle proliferation in perinatal cardiomyocytes. Although cardiomyocyte cell division and replication in mammals generally declines soon after birth, inhibition of ALMS1 expression in mice lead to increased cardiomyocyte proliferation, and deficiency of Alstrom protein has been suggested to impair post-natal cardiomyocyte cell cycle arrest. Here we describe the association of familial hypertrophic cardiomyopathy in Sphynx cats with a novel ALMS1 mutation. RESULTS A G/C variant was identified in exon 12 (human exon 13) of the ALMS1 gene in affected cats and was positively associated with the presence of hypertrophic cardiomyopathy in the feline population (p < 0.0001). The variant was predicted to change a highly conserved nonpolar Glycine to a positively charged Arginine. This was predicted to be a deleterious change by three in silico programs. Protein prediction programs indicated that the variant changed the protein structure in this region from a coil to a helix. Light microscopy findings included myofiber disarray with interstitial fibrosis with significantly more nuclear proliferative activity in the affected cats than controls (p < 0.0001). CONCLUSION This study demonstrates a novel form of cardiomyopathy associated with ALMS1 in the cat. Familial hypertrophic cardiomyopathy is a disease of genetic heterogeneity; many of the known causative genes encoding for sarcomeric proteins. Our findings suggest that variants in genes involved with cardiac development and cell regulation, like the ALMS1 gene, may deserve further consideration for association with familial hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Kathryn M Meurs
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA.
| | - Brian G Williams
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Dylan DeProspero
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, MN, 55108, USA
| | - David E Malarkey
- National Toxicology Program Pathology Group, Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - J Ashley Ezzell
- Histology Research Core Facility, Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Bruce W Keene
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Darcy B Adin
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Teresa C DeFrancesco
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Sandra Tou
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
29
|
Meurs KM, Williams BG, DeProspero D, Friedenberg SG, Malarkey DE, Ezzell JA, Keene BW, Adin DB, DeFrancesco TC, Tou S. A deleterious mutation in the ALMS1 gene in a naturally occurring model of hypertrophic cardiomyopathy in the Sphynx cat. Orphanet J Rare Dis 2021; 16:108. [PMID: 33639992 PMCID: PMC7913409 DOI: 10.1186/s13023-021-01740-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background Familial hypertrophic cardiomyopathy is a common inherited cardiovascular disorder in people. Many causal mutations have been identified, but about 40% of cases do not have a known causative mutation. Mutations in the ALMS1 gene are associated with the development of Alstrom syndrome, a multisystem familial disease that can include cardiomyopathy (dilated, restrictive). Hypertrophic cardiomyopathy has not been described. The ALMS1 gene is a large gene that encodes for a ubiquitously expressed protein. The function of the protein is not well understood although it is believed to be associated with energy metabolism and homeostasis, cell differentiation and cell cycle control. The ALMS1 protein has also been shown to be involved in the regulation of cell cycle proliferation in perinatal cardiomyocytes. Although cardiomyocyte cell division and replication in mammals generally declines soon after birth, inhibition of ALMS1 expression in mice lead to increased cardiomyocyte proliferation, and deficiency of Alstrom protein has been suggested to impair post-natal cardiomyocyte cell cycle arrest. Here we describe the association of familial hypertrophic cardiomyopathy in Sphynx cats with a novel ALMS1 mutation.
Results A G/C variant was identified in exon 12 (human exon 13) of the ALMS1 gene in affected cats and was positively associated with the presence of hypertrophic cardiomyopathy in the feline population (p < 0.0001). The variant was predicted to change a highly conserved nonpolar Glycine to a positively charged Arginine. This was predicted to be a deleterious change by three in silico programs. Protein prediction programs indicated that the variant changed the protein structure in this region from a coil to a helix. Light microscopy findings included myofiber disarray with interstitial fibrosis with significantly more nuclear proliferative activity in the affected cats than controls (p < 0.0001).
Conclusion This study demonstrates a novel form of cardiomyopathy associated with ALMS1 in the cat. Familial hypertrophic cardiomyopathy is a disease of genetic heterogeneity; many of the known causative genes encoding for sarcomeric proteins. Our findings suggest that variants in genes involved with cardiac development and cell regulation, like the ALMS1 gene, may deserve further consideration for association with familial hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Kathryn M Meurs
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA.
| | - Brian G Williams
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Dylan DeProspero
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, University of Minnesota, Saint Paul, MN, 55108, USA
| | - David E Malarkey
- National Toxicology Program Pathology Group, Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - J Ashley Ezzell
- Histology Research Core Facility, Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Bruce W Keene
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Darcy B Adin
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Teresa C DeFrancesco
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Sandra Tou
- Department of Veterinary Clinical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
30
|
Tallo CA, Duncan LH, Yamamoto AH, Slaydon JD, Arya GH, Turlapati L, Mackay TFC, Carbone MA. Heat shock proteins and small nucleolar RNAs are dysregulated in a Drosophila model for feline hypertrophic cardiomyopathy. G3 (BETHESDA, MD.) 2021; 11:jkaa014. [PMID: 33561224 PMCID: PMC7849908 DOI: 10.1093/g3journal/jkaa014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/16/2020] [Indexed: 11/13/2022]
Abstract
In cats, mutations in myosin binding protein C (encoded by the MYBPC3 gene) have been associated with hypertrophic cardiomyopathy (HCM). However, the molecular mechanisms linking these mutations to HCM remain unknown. Here, we establish Drosophila melanogaster as a model to understand this connection by generating flies harboring MYBPC3 missense mutations (A31P and R820W) associated with feline HCM. The A31P and R820W flies displayed cardiovascular defects in their heart rates and exercise endurance. We used RNA-seq to determine which processes are misregulated in the presence of mutant MYBPC3 alleles. Transcriptome analysis revealed significant downregulation of genes encoding small nucleolar RNA (snoRNAs) in exercised female flies harboring the mutant alleles compared to flies that harbor the wild-type allele. Other processes that were affected included the unfolded protein response and immune/defense responses. These data show that mutant MYBPC3 proteins have widespread effects on the transcriptome of co-regulated genes. Transcriptionally differentially expressed genes are also candidate genes for future evaluation as genetic modifiers of HCM as well as candidate genes for genotype by exercise environment interaction effects on the manifestation of HCM; in cats as well as humans.
Collapse
Affiliation(s)
- Christian A Tallo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Laura H Duncan
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Akihiko H Yamamoto
- The Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC 27695-7613, USA
| | - Joshua D Slaydon
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Gunjan H Arya
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Lavanya Turlapati
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Trudy F C Mackay
- The Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, Greenwood, SC 29646, USA
| | - Mary A Carbone
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695, USA
- The Center for Integrated Fungal Research and Department of Plant and Microbial Biology, North Carolina State University, Raleigh NC 27695-7244, USA
| |
Collapse
|
31
|
Tallo CA, Duncan LH, Yamamoto AH, Slaydon JD, Arya GH, Turlapati L, Mackay TFC, Carbone MA. Heat shock proteins and small nucleolar RNAs are dysregulated in a Drosophila model for feline hypertrophic cardiomyopathy. G3 (BETHESDA, MD.) 2021. [PMID: 33561224 DOI: 10.1093/g3journal/jkaa014.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In cats, mutations in myosin binding protein C (encoded by the MYBPC3 gene) have been associated with hypertrophic cardiomyopathy (HCM). However, the molecular mechanisms linking these mutations to HCM remain unknown. Here, we establish Drosophila melanogaster as a model to understand this connection by generating flies harboring MYBPC3 missense mutations (A31P and R820W) associated with feline HCM. The A31P and R820W flies displayed cardiovascular defects in their heart rates and exercise endurance. We used RNA-seq to determine which processes are misregulated in the presence of mutant MYBPC3 alleles. Transcriptome analysis revealed significant downregulation of genes encoding small nucleolar RNA (snoRNAs) in exercised female flies harboring the mutant alleles compared to flies that harbor the wild-type allele. Other processes that were affected included the unfolded protein response and immune/defense responses. These data show that mutant MYBPC3 proteins have widespread effects on the transcriptome of co-regulated genes. Transcriptionally differentially expressed genes are also candidate genes for future evaluation as genetic modifiers of HCM as well as candidate genes for genotype by exercise environment interaction effects on the manifestation of HCM; in cats as well as humans.
Collapse
Affiliation(s)
- Christian A Tallo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Laura H Duncan
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Akihiko H Yamamoto
- The Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC 27695-7613, USA
| | - Joshua D Slaydon
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Gunjan H Arya
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Lavanya Turlapati
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7614, USA
| | - Trudy F C Mackay
- The Center for Human Genetics and Department of Genetics and Biochemistry, Clemson University, Greenwood, SC 29646, USA
| | - Mary A Carbone
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695, USA.,The Center for Integrated Fungal Research and Department of Plant and Microbial Biology, North Carolina State University, Raleigh NC 27695-7244, USA
| |
Collapse
|
32
|
Ryser-Degiorgis MP, Robert N, Meier RK, Zürcher-Giovannini S, Pewsner M, Ryser A, Breitenmoser U, Kovacevic A, Origgi FC. Cardiomyopathy Associated With Coronary Arteriosclerosis in Free-Ranging Eurasian Lynx ( Lynx lynx carpathicus). Front Vet Sci 2020; 7:594952. [PMID: 33409296 PMCID: PMC7779598 DOI: 10.3389/fvets.2020.594952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/26/2020] [Indexed: 11/30/2022] Open
Abstract
The Eurasian lynx (subspecies Lynx lynx carpathicus) was reintroduced to Switzerland in the 1970's. Health monitoring of the reintroduced population started in the late 1980's. Since then, six lynx have been found affected by a myocardial disease. The earliest case was an animal that died after a field anesthesia. Two lynx were found dead, two were euthanized/culled because of disease signs, and one was hit by car. Two had a heart murmur at clinical examination. At necropsy, the first animal showed only lung edema but the other five had cardiomegaly associated with myocardial fibrosis. Three had multisystemic effusions. Histological examination of all six lynx showed mild to severe, multifocal, myocardial interstitial and perivascular fibrosis along with multifocal myocyte degeneration and loss, and replacement fibrosis. Moderate to severe multifocal arteriosclerosis with associated luminal stenosis of the small and medium-sized intramural coronary arteries and the presence of Anitschkow cells was also observed. The heart lesions may have led to sudden death in the first case and to a chronic right-sided heart failure in the remaining. None of the lynx showed lesions or signs suggestive of an acute or subacute infection. Given the common geographic origin of these animals and the severe loss of heterozygocity in this population, a genetic origin of the disease is hypothesized.
Collapse
Affiliation(s)
| | - Nadia Robert
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Roman Kaspar Meier
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Mirjam Pewsner
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | | | - Alan Kovacevic
- Small Animal Clinic, Department of Clinical Veterinary Science, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Francesco C Origgi
- Centre for Fish and Wildlife Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Buckley RM, Davis BW, Brashear WA, Farias FHG, Kuroki K, Graves T, Hillier LW, Kremitzki M, Li G, Middleton RP, Minx P, Tomlinson C, Lyons LA, Murphy WJ, Warren WC. A new domestic cat genome assembly based on long sequence reads empowers feline genomic medicine and identifies a novel gene for dwarfism. PLoS Genet 2020; 16:e1008926. [PMID: 33090996 PMCID: PMC7581003 DOI: 10.1371/journal.pgen.1008926] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 12/30/2022] Open
Abstract
The domestic cat (Felis catus) numbers over 94 million in the USA alone, occupies households as a companion animal, and, like humans, suffers from cancer and common and rare diseases. However, genome-wide sequence variant information is limited for this species. To empower trait analyses, a new cat genome reference assembly was developed from PacBio long sequence reads that significantly improve sequence representation and assembly contiguity. The whole genome sequences of 54 domestic cats were aligned to the reference to identify single nucleotide variants (SNVs) and structural variants (SVs). Across all cats, 16 SNVs predicted to have deleterious impacts and in a singleton state were identified as high priority candidates for causative mutations. One candidate was a stop gain in the tumor suppressor FBXW7. The SNV is found in cats segregating for feline mediastinal lymphoma and is a candidate for inherited cancer susceptibility. SV analysis revealed a complex deletion coupled with a nearby potential duplication event that was shared privately across three unrelated cats with dwarfism and is found within a known dwarfism associated region on cat chromosome B1. This SV interrupted UDP-glucose 6-dehydrogenase (UGDH), a gene involved in the biosynthesis of glycosaminoglycans. Importantly, UGDH has not yet been associated with human dwarfism and should be screened in undiagnosed patients. The new high-quality cat genome reference and the compilation of sequence variation demonstrate the importance of these resources when searching for disease causative alleles in the domestic cat and for identification of feline biomedical models. The practice of genomic medicine is predicated on the availability of a high quality reference genome and an understanding of the impact of genome variation. Such resources have lead to countless discoveries in humans, however by working exclusively within the framework of human genetics, our potential for understanding diseases biology is limited, as similar analyses in other species have often lead to novel insights. The generation of Felis_catus_9.0, a new high quality reference genome for the domestic cat, helps facilitate the expansion of genomic medicine into the Felis lineage. Using Felis_catus_9.0 we analyze the landscape of genomic variation from a collection of 54 cats within the context of human gene constraint. The distribution of variant impacts in cats is correlated with patterns of gene constraint in humans, indicating the utility of this reference for identifying novel mutations that cause phenotypes relevant to human and cat health. Moreover, structural variant analysis revealed a novel variant for feline dwarfism in UGDH, a gene that has not been associated with dwarfism in any other species, suggesting a role for UGDH in cases of undiagnosed dwarfism in humans.
Collapse
Affiliation(s)
- Reuben M. Buckley
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Brian W. Davis
- Department of Veterinary Integrative Biosciences, Interdisciplinary Program in Genetics, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Wesley A. Brashear
- Department of Veterinary Integrative Biosciences, Interdisciplinary Program in Genetics, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Fabiana H. G. Farias
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics, Washington University, St. Louis, Missouri, United States of America
| | - Kei Kuroki
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Tina Graves
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - LaDeana W. Hillier
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Milinn Kremitzki
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Gang Li
- Department of Veterinary Integrative Biosciences, Interdisciplinary Program in Genetics, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | | | - Patrick Minx
- Donald Danforth Plant Science, St Louis, Missouri, United States of America
| | - Chad Tomlinson
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Leslie A. Lyons
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - William J. Murphy
- Department of Veterinary Integrative Biosciences, Interdisciplinary Program in Genetics, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Wesley C. Warren
- Division of Animal Sciences, School of Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
34
|
Horowitz BN, Kutinsky IB, Linde A. Species-Spanning Echocardiography: Cardiovascular Insights from Across the Animal Kingdom. Curr Cardiol Rep 2020; 22:165. [PMID: 33037937 DOI: 10.1007/s11886-020-01417-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/03/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW The objective of this review is to present comparative echocardiography as a source of insights for human cardiovascular medicine. RECENT FINDINGS We present echocardiographic examples of high impact human cardiovascular pathologies, including valvular, vascular, conduction, and myocardial disorders, in a wide range of species in varying environments. Unique features associated with comparative echocardiographic assessments are linked to human cardiology, including natural animal models of resistance and vulnerability. The cardiovascular vulnerabilities and strengths of other species can be a source of invaluable insights for human healthcare professionals. Echocardiography is playing a key role in bridging human and veterinary cardiology. Consequently, species-spanning echocardiography can deliver novel insights for human medicine.
Collapse
Affiliation(s)
- B N Horowitz
- Department of Medicine, Harvard Medical School, Boston, MA, USA. .,Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA. .,David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Ilana B Kutinsky
- William Beaumont School of Medicine, Oakland University, Rochester, MI, USA
| | - Annika Linde
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
35
|
Oldt RF, Bussey KJ, Settles ML, Fass JN, Roberts JA, Reader JR, Komandoor S, Abrich VA, Kanthaswamy S. MYBPC3 Haplotype Linked to Hypertrophic Cardiomyopathy in Rhesus Macaques ( Macaca mulatta). Comp Med 2020; 70:358-367. [PMID: 32753092 PMCID: PMC7574221 DOI: 10.30802/aalas-cm-19-000108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/13/2020] [Accepted: 02/07/2020] [Indexed: 11/05/2022]
Abstract
In humans, abnormal thickening of the left ventricle of the heart clinically defines hypertrophic cardiomyopathy (HCM), a common inherited cardiovascular disorder that can precede a sudden cardiac death event. The wide range of clinical presentations in HCM obscures genetic variants that may influence an individual's susceptibility to sudden cardiac death. Although exon sequencing of major sarcomere genes can be used to detect high-impact causal mutations, this strategy is successful in only half of patient cases. The incidence of left ventricular hypertrophy (LVH) in a managed research colony of rhesus macaques provides an excellent comparative model in which to explore the genomic etiology of severe HCM and sudden cardiac death. Because no rhesus HCM-associated mutations have been reported, we used a next-generation genotyping assay that targets 7 sarcomeric rhesus genes within 63 genomic sites that are orthologous to human genomic regions known to harbor HCM disease variants. Amplicon sequencing was performed on 52 macaques with confirmed LVH and 42 unrelated, unaffected animals representing both the Indian and Chinese rhesus macaque subspecies. Bias-reduced logistic regression uncovered a risk haplotype in the rhesus MYBPC3 gene, which is frequently disrupted in both human and feline HCM; this haplotype implicates an intronic variant strongly associated with disease in either homozygous or carrier form. Our results highlight that leveraging evolutionary genomic data provides a unique, practical strategy for minimizing population bias in complex disease studies.
Collapse
Affiliation(s)
- Robert F Oldt
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; Evolutionary Biology Graduate Program, School of Life Sciences, Arizona State University at the West Campus, Glendale, Arizona;,
| | - Kimberly J Bussey
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; BEYOND Center for Fundamental Concepts in Science, Arizona State University at the West Campus, Glendale, Arizona
| | - Matthew L Settles
- Bioinformatics Core, UC Davis Genome Center, University of California, Davis, California
| | - Joseph N Fass
- Bioinformatics Core, UC Davis Genome Center, University of California, Davis, California
| | - Jeffrey A Roberts
- California National Primate Research Center, University of California, Davis, California
| | - J Rachel Reader
- California National Primate Research Center, University of California, Davis, California
| | | | - Victor A Abrich
- Division of Cardiovascular Diseases, Mayo Clinic, Scottsdale, Arizona
| | - Sreetharan Kanthaswamy
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; Evolutionary Biology Graduate Program, School of Life Sciences, Arizona State University at the West Campus, Glendale, Arizona; California National Primate Research Center, University of California, Davis, California
| |
Collapse
|
36
|
Liu M, Eckersall PD, Mrljak V, Horvatić A, Guillemin N, Galan A, Köster L, French A. Novel biomarkers in cats with congestive heart failure due to primary cardiomyopathy. J Proteomics 2020; 226:103896. [PMID: 32652222 DOI: 10.1016/j.jprot.2020.103896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/10/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022]
Abstract
The pathogenesis of feline cardiomyopathy and congestive heart failure (CHF) requires further understanding. In this study, we assessed serum proteome change in feline CHF, aiming to identify novel biomarker for both research and clinical use. The study comprised 15 cats in CHF, 5 cats in preclinical cardiomyopathy and 15 cats as healthy controls. Serum proteome profiles were obtained by tandem mass tag labelling followed by mass spectrometry. Protein concentrations in CHF cats were compared with healthy controls. Western blot was performed for proteomic validation. Correlations were assessed between the altered proteins in CHF and clinical variables in cats with cardiomyopathy to evaluate protein-cardiac association. Bioinformatic analysis was employed to identify pathophysiological pathways involved in feline CHF. Sixteen serum proteins were significantly different between CHF and healthy control cats (P < .05). These included serine protease inhibitors, apolipoproteins and other proteins associated with inflammation and coagulation. Clinical parameters from cats with cardiomyopathy significantly correlated with the altered proteins (P < .05). Bioinformatic analysis identified 13 most relevant functional profiles in feline CHF, which mostly associated with extracellular matrix organization and metabolism. Data are available via ProteomeXchange with identifier PXD017761. SIGNIFICANCE: Cardiomyopathies affect both cats and humans, and they can cause serious consequence such as congestive heart failure (CHF). To date, the pathophysiological mechanism of CHF is not fully understood. In this study, for the first time, we used a proteomic approach combined with bioinformatic analysis to evaluate serum protein change in cats with CHF. Results indicate systemic inflammation, coagulation protein changes, innate immunity and extracellular matrix remodeling are involved in feline CHF, which are largely comparable with findings in previous human studies. Our study provides new insights into CHF and cardiomyopathy in cats, and the identified novel biomarkers and pathophysiological pathways provide valuable information for future studies.
Collapse
Affiliation(s)
- Mengmeng Liu
- Small Animal Hospital, School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - P David Eckersall
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK; Laboratory for Proteomics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Vladimir Mrljak
- Laboratory for Proteomics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Anita Horvatić
- Laboratory for Proteomics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Nicolas Guillemin
- Laboratory for Proteomics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Asier Galan
- Laboratory for Proteomics, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Liza Köster
- Department of Small Animal Clinical Sciences, University of Tennessee, Knoxville, TN, USA
| | - Anne French
- Ross University School of Veterinary Medicine, Basseterre, St Kitts & Nevis, West Indies.
| |
Collapse
|
37
|
Gil-Ortuño C, Sebastián-Marcos P, Sabater-Molina M, Nicolas-Rocamora E, Gimeno-Blanes JR, Fernández Del Palacio MJ. Genetics of feline hypertrophic cardiomyopathy. Clin Genet 2020; 98:203-214. [PMID: 32215921 DOI: 10.1111/cge.13743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/29/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by an abnormal increase in myocardial mass that affects cardiac structure and function. HCM is the most common inherited cardiovascular disease in humans (0.2%) and the most common cardiovascular disease in cats (14.7%). Feline HCM phenotype is very similar to the phenotype found in humans, but the time frame for the development of the disease is significantly shorter. Similar therapeutic agents are used in its treatment and it has the same complications, such as heart failure, thromboembolism and sudden cardiac death. In contrast to humans, in whom thousands of genetic variants have been identified, genetic studies in cats have been limited to fragment analysis of two sarcomeric genes identifying two variants in MYBPC3 and one in MYH7. Two of these variants have also been associated with human disease. The high prevalence of the reported variants in non-affected cats hinders the assumption of their pathogenicity in heterozygotes. An in-depth review of the literature about genetic studies on feline HCM in comparison with the same disease in humans is presented here. The close similarity in the phenotype and genotype between cats and humans makes the cat an excellent model for the pathophysiological study of the disease and future therapeutic agents.
Collapse
Affiliation(s)
- Cristina Gil-Ortuño
- Cardiogenetic Laboratory, Inherited Cardiac Disease Unit, IMIB University Hospital Virgen de la Arrixaca-IMIB, Murcia, Spain
| | | | - María Sabater-Molina
- Cardiogenetic Laboratory, Inherited Cardiac Disease Unit, IMIB University Hospital Virgen de la Arrixaca-IMIB, Murcia, Spain.,Internal Medicine Department, University of Murcia, Murcia, Spain
| | - Elisa Nicolas-Rocamora
- Cardiogenetic Laboratory, Inherited Cardiac Disease Unit, IMIB University Hospital Virgen de la Arrixaca-IMIB, Murcia, Spain
| | - Juan R Gimeno-Blanes
- Internal Medicine Department, University of Murcia, Murcia, Spain.,Department of Cardiology, Inherited Cardiac Disease Unit, University Hospital Virgen de la Arrixaca, Murcia, Spain
| | | |
Collapse
|
38
|
DISEASE AND PATHOLOGICAL CONDITIONS OF AN ENDANGERED RODENT, MICROTUS CALIFORNICUS SCIRPENSIS, IN A CAPTIVE-REARING FACILITY AND IN THE WILD. J Zoo Wildl Med 2020; 50:758-768. [PMID: 31926505 DOI: 10.1638/2018-0117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2019] [Indexed: 11/21/2022] Open
Abstract
Causes of morbidity and mortality and a survey of infectious disease agents were collated from wild and colony-raised endangered Amargosa voles (Microtus californicus scirpensis). Six voles from the wild and 295 voles in the captive-breeding colony were included in the study upon identification of an infectious agent during screening, identification of clinical signs of disease, or finding a pathological condition or infectious agent on necropsy. Findings included 28 significant or incidental pathological conditions of seven organ systems and 19 parasitic, viral, bacterial, or fungal agents. Several voles captured in the wild had fungal osteomyelitis of the tail that disseminated systemically in a vole brought from the wild to the colony and may have been caused by a Penicillium sp. Three voles reintroduced from the colony to the wild experienced inanition and subsequent severe hepatic and moderate renal tubular lipidosis. The most common significant pathological conditions in colony-reared voles were chronic interstitial nephritis with proteinosis; cardiomyopathy; trichobezoars that, in intestines or cecocolic junctions, sometimes induced local rupture or infarction with peritonitis; multifocal gastrointestinal ulceration and colibacillosis; acute renal tubular necrosis or nephritis; sepsis; hepatic and renal lipidosis; molar apical elongation sometimes progressing to invasion of the calvarium; and mammary tumors. Uncommon diagnoses included intervertebral disc disease; microvascular dysplasia; and multifocal bacterial abscessation. Common or clinically important infectious agents included Demodex sp. mites in hair follicles, Demodex sp. in esophageal mucosa, and an outbreak of tropical rat mites thought to have been introduced via the straw bedding; gastrointestinal Helicobacter sp.; attaching and effacing Escherichia coli; and Citrobacter braakii, a possible zoonotic bacterium. This survey of species-specific diseases and pathogens was possible because the established health surveillance program that is part of the species recovery plan allowed for monitoring of voles throughout the duration of their natural life spans in captivity.
Collapse
|
39
|
Du Y, Wang Y, Han X, Feng Z, Ma A. MYH7 Gene-Related Mutation p.V878L Identified in a Chinese Family with Hypertrophic Cardiomyopathy. Int Heart J 2019; 60:1415-1420. [PMID: 31735781 DOI: 10.1536/ihj.19-146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common inherited cardiovascular diseases and possesses a high risk for sudden cardiac death. Although mutations in more than 20 genes have been reported to be associated with HCM thus far, the genetic backgrounds of most HCM patients are not fully understood. We performed a genetic analysis in a Chinese family that presented with HCM using next-generation sequencing (NGS). Clinical data, family histories, and blood samples were collected from the proband and family members. Five patients showed typical clinical symptoms of HCM. One subject was the victim of sudden cardiac death. By NGS, we determined that these subjects with HCM symptoms carried a missense heterozygous genetic mutation c.2632C>A (p.V878L) in the myosin heavy chain 7 (MYH7) gene with an autosomal dominant pattern of inheritance. Individuals without this mutation showed no symptoms or cardiac structural abnormalities related to HCM. Bioinformatics evaluation predicted this mutant as "damaging" and "disease causing". Additionally, sequence alignment showed that this mutant is located in an evolutionarily conserved region of MYH7 in multiple species. Our results describe a potentially pathogenic mutation associated with HCM, which may extend the spectrum of HCM phenotypes related to MYH7 gene mutations.
Collapse
Affiliation(s)
- Yuan Du
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University
| | - Ya Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University
| | - Xiu Han
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University
| | - Zhanbin Feng
- Department of Cardiovascular Medicine, Ninth Hospital of Xi'an
| | - Aiqun Ma
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University.,Shaanxi Key Laboratory of Molecular Cardiology, Xi'an Jiaotong University.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education
| |
Collapse
|
40
|
Stern JA, Ueda Y. Inherited cardiomyopathies in veterinary medicine. Pflugers Arch 2018; 471:745-753. [PMID: 30284024 DOI: 10.1007/s00424-018-2209-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Abstract
Comparative and translation medicine is of particular value within the field of inherited cardiomyopathies. Despite massive advances in understanding the functional role of mutations in human cardiomyopathies, these advances have frequently failed to translate into medical discoveries that alter patient care. One potential explanation for this failure lies in the lack of suitable translational models that adequately recapitulate human cardiovascular physiology and disease expression. The vast genetic heterogeneity that complicates human cardiomyopathy research is potentially alleviated through the study of naturally occurring large animal models of disease, where incredibly homogenous populations, like those seen in a single breed of dog or cat, may exist (Kol et al., Sci Transl Med 7:308-321, 2015; Ueda and Stern, Yale J Biol Med 90:433-448, 2017). Veterinary medicine is in a unique position to provide research resources and information that may be readily applied to human disease (Kol et al., Sci Transl Med 7:308-321, 2015). Many inherited cardiomyopathies of humans are phenotypically and genotypically similar in veterinary species and ongoing research holds promise for aiding veterinary and human patients alike (Basso et al., Circulation 109:1180-1185, 2004; Fox et al., Cardiovasc Pathol 23:28-34, 2014; Fox et al., Circulation 102:1863-1870, 2000; Kittleson et al., J Vet Cardiol 17 Suppl 1:S53-73, 2015; Ueda and Stern, Yale J Biol Med 90:433-448, 2017). This article presents the current knowledge of inherited cardiomyopathies in dogs, cats, and non-human primates, with a goal of identifying areas of translational research and future directions.
Collapse
Affiliation(s)
- Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA.
| | - Yu Ueda
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, One Shields Avenue, Davis, CA, USA
| |
Collapse
|
41
|
Miller DL, Lu X, Dou C, Zhu YI, Fuller R, Fields K, Fabiilli ML, Owens GE, Gordon D, Kripfgans OD. Ultrasonic Cavitation-Enabled Treatment for Therapy of Hypertrophic Cardiomyopathy: Proof of Principle. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1439-1450. [PMID: 29681423 PMCID: PMC5960614 DOI: 10.1016/j.ultrasmedbio.2018.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 05/08/2023]
Abstract
Ultrasound myocardial cavitation-enabled treatment was applied to the SS-16BN rat model of hypertrophic cardiomyopathy for proof of the principle underlying myocardial reduction therapy. A focused ultrasound transducer was targeted using 10-MHz imaging (10 S, GE Vivid 7) to the left ventricular wall of anesthetized rats in a warmed water bath. Pulse bursts of 4-MPa peak rarefactional pressure amplitude were intermittently triggered 1:8 heartbeats during a 10-min infusion of a microbubble suspension. Methylprednisolone was given to reduce initial inflammation, and Losartan was given to reduce fibrosis in the healing tissue. At 28 d post therapy, myocardial cavitation-enabled treatment significantly reduced the targeted wall thickness by 16.2% (p <0.01) relative to shams, with myocardial strain rate and endocardial displacement reduced by 34% and 29%, respectively, which are sufficient for therapeutic treatment. Premature electrocardiogram complexes and plasma troponin measurements were found to identify optimal and suboptimal treatment cohorts and would aid in achieving the desired impact. With clinical translation, myocardial cavitation-enabled treatment should fill the need for a new non-invasive hypertrophic cardiomyopathy therapy option.
Collapse
Affiliation(s)
| | - Xiaofang Lu
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Chunyan Dou
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Yiying I Zhu
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Rachael Fuller
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Kristina Fields
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | | | - Gabe E Owens
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | - David Gordon
- University of Michigan Health System, Ann Arbor, Michigan, USA
| | | |
Collapse
|
42
|
Wang J, Zhang X, Wang X, Wang C, Wang F, Wang B. MYH7 Rare Variant in a Family With Double-Chambered Left Ventricle. ACTA ACUST UNITED AC 2017; 10:CIRCGENETICS.117.001729. [PMID: 29237678 DOI: 10.1161/circgenetics.117.001729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jing Wang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.)
| | - Xin Zhang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.)
| | - Xi Wang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.)
| | - Chuchu Wang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.)
| | - Fangyun Wang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.).
| | - Binbin Wang
- From the Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences (J.W.) and Heart Center, Beijing Children's Hospital (X.Z., F.W.), Capital Medical University, China; Center for Genetics, National Research Institute for Family Planning, Beijing (X.W., B.W.); and School of Life Sciences, Zhengzhou University, China (C.W.).
| |
Collapse
|
43
|
Ueda Y, Stern JA. A One Health Approach to Hypertrophic Cardiomyopathy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:433-448. [PMID: 28955182 PMCID: PMC5612186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease in humans and results in significant morbidity and mortality. Research over the past 25 years has contributed enormous insight into this inherited disease particularly in the areas of genetics, molecular mechanisms, and pathophysiology. Our understanding continues to be limited by the heterogeneity of clinical presentations with various genetic mutations associated with HCM. Transgenic mouse models have been utilized especially studying the genotypic and phenotypic interactions. However, mice possess intrinsic cardiac and hemodynamic differences compared to humans and have limitations preventing their direct translation. Other animal models of HCM have been studied or generated in part to overcome these limitations. HCM in cats shows strikingly similar molecular, histopathological, and genetic similarities to human HCM, and offers an important translational opportunity for the study of this disease. Recently, inherited left ventricular hypertrophy in rhesus macaques was identified and collaborative investigations have been conducted to begin to develop a non-human primate HCM model. These naturally-occurring large-animal models may aid in advancing our understanding of HCM and developing novel therapeutic approaches to this disease. This review will highlight the features of HCM in humans and the relevant available and developing animal models of this condition.
Collapse
Affiliation(s)
- Yu Ueda
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA
| | - Joshua A. Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis, CA,California National Primate Research Center, University of California-Davis, Davis, CA,To whom all correspondence should be addressed: Joshua A. Stern, One Shields Avenue, Davis, CA, 95616, Tel: 530-752-2475, .
| |
Collapse
|
44
|
Freeman LM, Rush JE, Stern JA, Huggins GS, Maron MS. Feline Hypertrophic Cardiomyopathy: A Spontaneous Large Animal Model of Human HCM. Cardiol Res 2017; 8:139-142. [PMID: 28868097 PMCID: PMC5574284 DOI: 10.14740/cr578w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/28/2017] [Indexed: 11/25/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a common disease in pet cats, affecting 10-15% of the pet cat population. The similarity to human HCM, the rapid progression of disease, and the defined and readily determined endpoints of feline HCM make it an excellent natural model that is genotypically and phenotypically similar to human HCM. The Maine Coon and Ragdoll cats are particularly valuable models of HCM because of myosin binding protein-C mutations and even higher disease incidence compared to the overall feline population. The cat overcomes many of the limitations of rodent HCM models, and can provide enhanced translation of information from in vitro and induced small animal models to human clinical trials. Physicians and veterinarians working together in a collaborative and interdisciplinary approach can accelerate the discovery of more effective treatments for this and other cardiovascular diseases affecting human and veterinary patients.
Collapse
Affiliation(s)
- Lisa M Freeman
- Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, USA
| | - John E Rush
- Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, 200 Westboro Road, North Grafton, MA, USA
| | - Joshua A Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, CCAH Room 258, One Shields Avenue, Davis, CA, USA
| | - Gordon S Huggins
- Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Martin S Maron
- Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| |
Collapse
|
45
|
Fox PR, Kittleson MD, Basso C, Thiene G. Letter to the Editor. J Vet Intern Med 2017. [PMID: 28626958 PMCID: PMC5508327 DOI: 10.1111/jvim.14780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- P R Fox
- Caspary Research Institute of the Animal Medical Center, New York, NY
| | - M D Kittleson
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA
| | - C Basso
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padova, Italy
| | - G Thiene
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua Medical School, Padova, Italy
| |
Collapse
|
46
|
A Small Molecule Inhibitor of Sarcomere Contractility Acutely Relieves Left Ventricular Outflow Tract Obstruction in Feline Hypertrophic Cardiomyopathy. PLoS One 2016. [PMID: 27973580 DOI: 10.1371/journal.pone.0168407.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disease of the heart muscle characterized by otherwise unexplained thickening of the left ventricle. Left ventricular outflow tract (LVOT) obstruction is present in approximately two-thirds of patients and substantially increases the risk of disease complications. Invasive treatment with septal myectomy or alcohol septal ablation can improve symptoms and functional status, but currently available drugs for reducing obstruction have pleiotropic effects and variable therapeutic responses. New medical treatments with more targeted pharmacology are needed, but the lack of preclinical animal models for HCM with LVOT obstruction has limited their development. HCM is a common cause of heart failure in cats, and a subset exhibit systolic anterior motion of the mitral valve leading to LVOT obstruction. MYK-461 is a recently-described, mechanistically novel small molecule that acts at the sarcomere to specifically inhibit contractility that has been proposed as a treatment for HCM. Here, we use MYK-461 to test whether direct reduction in contractility is sufficient to relieve LVOT obstruction in feline HCM. We evaluated mixed-breed cats in a research colony derived from a Maine Coon/mixed-breed founder with naturally-occurring HCM. By echocardiography, we identified five cats that developed systolic anterior motion of the mitral valve and LVOT obstruction both at rest and under anesthesia when provoked with an adrenergic agonist. An IV MYK-461 infusion and echocardiography protocol was developed to serially assess contractility and LVOT gradient at multiple MYK-461 concentrations. Treatment with MYK-461 reduced contractility, eliminated systolic anterior motion of the mitral valve and relieved LVOT pressure gradients in an exposure-dependent manner. Our findings provide proof of principle that acute reduction in contractility with MYK-461 is sufficient to relieve LVOT obstruction. Further, these studies suggest that feline HCM will be a valuable translational model for the study of disease pathology, particularly LVOT obstruction.
Collapse
|
47
|
Stern JA, Markova S, Ueda Y, Kim JB, Pascoe PJ, Evanchik MJ, Green EM, Harris SP. A Small Molecule Inhibitor of Sarcomere Contractility Acutely Relieves Left Ventricular Outflow Tract Obstruction in Feline Hypertrophic Cardiomyopathy. PLoS One 2016; 11:e0168407. [PMID: 27973580 PMCID: PMC5156432 DOI: 10.1371/journal.pone.0168407] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/29/2016] [Indexed: 11/19/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an inherited disease of the heart muscle characterized by otherwise unexplained thickening of the left ventricle. Left ventricular outflow tract (LVOT) obstruction is present in approximately two-thirds of patients and substantially increases the risk of disease complications. Invasive treatment with septal myectomy or alcohol septal ablation can improve symptoms and functional status, but currently available drugs for reducing obstruction have pleiotropic effects and variable therapeutic responses. New medical treatments with more targeted pharmacology are needed, but the lack of preclinical animal models for HCM with LVOT obstruction has limited their development. HCM is a common cause of heart failure in cats, and a subset exhibit systolic anterior motion of the mitral valve leading to LVOT obstruction. MYK-461 is a recently-described, mechanistically novel small molecule that acts at the sarcomere to specifically inhibit contractility that has been proposed as a treatment for HCM. Here, we use MYK-461 to test whether direct reduction in contractility is sufficient to relieve LVOT obstruction in feline HCM. We evaluated mixed-breed cats in a research colony derived from a Maine Coon/mixed-breed founder with naturally-occurring HCM. By echocardiography, we identified five cats that developed systolic anterior motion of the mitral valve and LVOT obstruction both at rest and under anesthesia when provoked with an adrenergic agonist. An IV MYK-461 infusion and echocardiography protocol was developed to serially assess contractility and LVOT gradient at multiple MYK-461 concentrations. Treatment with MYK-461 reduced contractility, eliminated systolic anterior motion of the mitral valve and relieved LVOT pressure gradients in an exposure-dependent manner. Our findings provide proof of principle that acute reduction in contractility with MYK-461 is sufficient to relieve LVOT obstruction. Further, these studies suggest that feline HCM will be a valuable translational model for the study of disease pathology, particularly LVOT obstruction.
Collapse
Affiliation(s)
- Joshua A. Stern
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Svetlana Markova
- MyoKardia, Inc., South San Francisco, California, United States of America
| | - Yu Ueda
- Department of Medicine & Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Jae B. Kim
- MyoKardia, Inc., South San Francisco, California, United States of America
| | - Peter J. Pascoe
- Department of Surgical & Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Marc J. Evanchik
- MyoKardia, Inc., South San Francisco, California, United States of America
| | - Eric M. Green
- MyoKardia, Inc., South San Francisco, California, United States of America
| | - Samantha P. Harris
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|