1
|
Budiono BP, Vider J, Zaid A, Peart JN, Du Toit EF, Headrick JP, Haseler LJ. Swimming induces physiological cardioprotection associated with pro-growth versus anti-inflammatory influences in extracardiac organs. Am J Physiol Regul Integr Comp Physiol 2025; 328:R206-R219. [PMID: 39792091 DOI: 10.1152/ajpregu.00139.2024] [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: 05/22/2024] [Revised: 12/11/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
Physical activity improves myocardial structure, function, and resilience via complex, incompletely defined mechanisms. We explored the effects of 1- to 2-wk swim training on cardiac and systemic phenotype in young male C57Bl/6 mice. Two-week forced swimming (90 min twice daily) resulted in cardiac hypertrophy (22% increase in heart:body weight, P < 0.01), with improved inotropy (22% higher left ventricular +dP/dt, P < 0.01) and functional tolerance to ischemia-reperfusion (I-R) (40%-50% reductions in stunning and diastolic dysfunction, P < 0.01; without changes in cell death assessed from enzyme loss) in Langendorff perfused hearts. Initial Western immunoblot analysis indicated no shifts in cardiac expression of determinants of autophagy (LC3A/B), mitochondrial biogenesis/dynamics (PGC-1α, MFN-1, and OPA-1), or stress signaling (caveolin-3 and GSK-3β). Furthermore, no changes in cardiac cytokines (IL-1b, IL-6, IL-10, IL-12, GM-CSF, TNF-α, and IFN-γ) were detected in multiplex immunoassays. Exploratory profiling of RTK phosphorylation provided evidence for moderately increased activity of receptors involved in cardiac/coronary growth and protection (insulin, IGF-1, FGF R2, Tie-2, PDGFβ, and EphB4), together with a fall in M-CSF R and ephrin sub-type receptor phosphorylation. Swimming increased growth factor while reducing inflammatory mediators across extracardiac tissues [brain, pancreas, thymus, lymph nodes, and white adipose tissue (WAT)]. This included a pattern of increased LIF, VEGF, and pentraxin-2 versus reduced CXCL2/MIP-2a, chitinase 3-like 1, CCL6, MMP9, CD40/TNFRSF5, and IGFBP6 in multiple tissues, and a shift to a pro-browning profile in WAT. In summary, swimming produces integrated systemic benefits, improving cardiac growth, inotropy, and resilience in association with increased growth factor and reduced inflammatory and lipogenic mediators in multiple tissues.NEW & NOTEWORTHY Swimming may induce cardiac and systemic benefits distinct from other modes of physical activity. We show that 2-wk forced swim training increases cardiac growth, contractility, and functional resilience to ischemia in hearts of male mice. This is associated with increased growth factor levels and reduced inflammatory and lipogenic protein profiles in peripheral tissues.
Collapse
Affiliation(s)
- Boris P Budiono
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Jelena Vider
- School of Pharmacy and Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Ali Zaid
- School of Pharmacy and Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Jason N Peart
- School of Pharmacy and Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Eugene F Du Toit
- School of Pharmacy and Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - John P Headrick
- School of Pharmacy and Medical Science, Griffith University Gold Coast, Southport, Queensland, Australia
| | - Luke J Haseler
- Curtin School of Allied Health, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
2
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. N-Terminomic Identification of Intracellular MMP-2 Substrates in Cardiac Tissue. J Proteome Res 2024; 23:4188-4202. [PMID: 38647137 PMCID: PMC11460328 DOI: 10.1021/acs.jproteome.3c00755] [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: 11/11/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Proteases are enzymes that induce irreversible post-translational modifications by hydrolyzing amide bonds in proteins. One of these proteases is matrix metalloproteinase-2 (MMP-2), which has been shown to modulate extracellular matrix remodeling and intracellular proteolysis during myocardial injury. However, the substrates of MMP-2 in heart tissue are limited, and lesser known are the cleavage sites. Here, we used degradomics to investigate the substrates of intracellular MMP-2 in rat ventricular extracts. First, we designed a novel, constitutively active MMP-2 fusion protein (MMP-2-Fc) that we expressed and purified from mammalian cells. Using this protease, we proteolyzed ventricular extracts and used subtiligase-mediated N-terminomic labeling which identified 95 putative MMP-2-Fc proteolytic cleavage sites using mass spectrometry. The intracellular MMP-2 cleavage sites identified in heart tissue extracts were enriched for proteins primarily involved in metabolism, as well as the breakdown of fatty acids and amino acids. We further characterized the cleavage of three of these MMP-2-Fc substrates based on the gene ontology analysis. We first characterized the cleavage of sarco/endoplasmic reticulum calcium ATPase (SERCA2a), a known MMP-2 substrate in myocardial injury. We then characterized the cleavage of malate dehydrogenase (MDHM) and phosphoglycerate kinase 1 (PGK1), representing new cardiac tissue substrates. Our findings provide insights into the intracellular substrates of MMP-2 in cardiac cells, suggesting that MMP-2 activation plays a role in cardiac metabolism.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
3
|
Dorey ES, Headrick JP, Paravicini TM, Wlodek ME, Moritz KM, Reichelt ME. Periconceptional alcohol alters in vivo heart function in ageing female rat offspring: Possible involvement of oestrogen receptor signalling. Exp Physiol 2023; 108:772-784. [PMID: 36951040 PMCID: PMC10988452 DOI: 10.1113/ep090587] [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: 06/13/2022] [Accepted: 02/22/2023] [Indexed: 03/24/2023]
Abstract
NEW FINDINGS What is the central question of this study? What are the cardiovascular consequences of periconceptual ethanol on offspring throughout the lifespan? What is the main finding and its importance? It is shown for the first time that periconceptional alcohol has sex-specific effects on heart growth, with ageing female offspring exhibiting decreased cardiac output. Altered in vivo cardiac function in ageing female offspring may be linked to changes in cardiac oestrogen receptor expression. ABSTRACT Alcohol exposure throughout gestation is detrimental to cardiac development and function. Although many women decrease alcohol consumption once aware of a pregnancy, exposure prior to recognition is common. We, therefore, examined the effects of periconceptional alcohol exposure (PC:EtOH) on heart function, and explored mechanisms that may contribute. Female Sprague-Dawley rats received a liquid diet ±12.5% v/v ethanol from 4 days prior to mating until 4 days after mating (PC:EtOH). Cardiac function was assessed via echocardiography, and offspring were culled at multiple time points for assessment of morphometry, isolated heart and aortic ring function, protein and transcriptional changes. PC:EtOH-exposed embryonic day 20 fetuses (but not postnatal offspring) had larger hearts relative to body weight. Ex vivo analysis of hearts at 5-7 months old (mo) indicated no changes in coronary function or cardiac ischaemic tolerance, and apparently improved ventricular compliance in PC:EtOH females (compared to controls). At 12 mo, vascular responses in isolated aortic rings were unaltered by PC:EtOH, whilst echocardiography revealed reduced cardiac output in female but not male PC:EtOH offspring. At 19 mo, left ventricular transcript and protein for type 1 oestrogen receptor (ESR1), HSP90 transcript and plasma oestradiol levels were all elevated in female PC:EtOH exposed offspring. Summarising, PC:EtOH adversely impacts in vivo heart function in mature female offspring, associated with increased ventricular oestrogen-related genes. PC:EtOH may thus influence age-related heart dysfunction in females through modulation of oestrogen signalling.
Collapse
Affiliation(s)
- Emily S. Dorey
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| | - John P. Headrick
- School of Pharmacy and Medical ScienceGriffith UniversitySouthportQueenslandAustralia
| | - Tamara M. Paravicini
- School of Health and Biomedical SciencesRMIT UniversityMelbourneVictoriaAustralia
| | - Mary E. Wlodek
- The Department of Obstetrics and GynaecologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Karen M. Moritz
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
- Child Health Research CentreUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Melissa E. Reichelt
- School of Biomedical SciencesUniversity of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
4
|
Ashton KJ, Kiessling CJ, Thompson JLM, Aziz AY, Thomas WG, Headrick JP, Reichelt ME. Early cardiac aging linked to impaired stress-resistance and transcriptional control of stress response, quality control and mitochondrial pathways. Exp Gerontol 2023; 171:112011. [PMID: 36347360 DOI: 10.1016/j.exger.2022.112011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Phenotypic and transcriptomic evidence of early cardiac aging, and associated mechanisms, were investigated in young to middle-aged male mice (C57Bl/6; ages 8, 16, 32, 48 wks). Left ventricular gene expression (profiled via Illumina MouseWG-6 BeadChips), contractile and coronary function, and stress-resistance were assessed in Langendorff perfused hearts under normoxic conditions and following ischemic insult (20 min global ischemia-45 min reperfusion; I-R). Baseline or normoxic contractile function was unaltered by age, while cardiac and coronary 'reserves' (during β-adrenoceptor stimulation; 1 μM isoproterenol) declined by 48 wks. Resistance to I-R injury fell from 16 to 32 wks. Age-dependent transcriptional changes In un-stressed hearts were limited to 104 genes (>1.3-fold; 0.05 FDR), supporting: up-regulated innate defenses (glutathione and xenobiotic metabolism, chemotaxis, interleukins) and catecholamine secretion; and down-regulated extracellular matrix (ECM), growth factor and survival (PI3K/Akt) signaling. In stressed (post-ischemic) myocardium, ∼15-times as many genes (1528) were age-dependent, grouped into 6 clusters (>1.3-fold change; 0.05 FDR): most changing from 16 wks (45 % up/44 % down), a further 5 % declining from 32 wks. Major age-dependent Biological Processes in I-R hearts reveal: declining ATP metabolism, oxidative phosphorylation, cardiac contraction and morphogenesis, phospholipid metabolism and calcineurin signaling; increasing proteolysis and negative control of MAPK; and mixed changes in nuclear transport and angiogenic genes. Pathway analysis supports reductions in: autophagy, stress response, ER protein processing, mRNA surveillance and ribosome/translation genes; with later falls in mitochondrial biogenesis, oxidative phosphorylation and proteasome genes in I-R hearts. Summarizing, early cardiac aging is evident from 16 to 32 wks in male mice, characterized by: declining cardiovascular reserve and stress-resistance, transcriptomic evidence of constitutive stress and altered catecholamine and survival/growth signaling in healthy hearts; and declining stress response, quality control, mitochondrial energy metabolism and cardiac modeling processes in stressed hearts. These very early changes, potentially key substrate for advanced aging, may inform approaches to healthy aging and cardioprotection in the adult heart.
Collapse
Affiliation(s)
- Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Can J Kiessling
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Jamie-Lee M Thompson
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Aliah Y Aziz
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - John P Headrick
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Zhao X, Yang X, An Z, Liu L, Yong J, Xing H, Huang R, Tian J, Song X. Pathophysiology and molecular mechanism of caveolin involved in myocardial protection strategies in ischemic conditioning. Biomed Pharmacother 2022; 153:113282. [PMID: 35750009 DOI: 10.1016/j.biopha.2022.113282] [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: 04/27/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022] Open
Abstract
Multiple pathophysiological pathways are activated during the process of myocardial injury. Various cardioprotective strategies protect the myocardium from ischemia, infarction, and ischemia/reperfusion (I/R) injury through different targets, yet the clinical translation remains limited. Caveolae and its structure protein, caveolins, have been suggested as a bridge to transmit damage-preventing signals and mediate the protection of ultrastructure in cardiomyocytes under pathological conditions. In this review, we first briefly introduce caveolae and caveolins. Then we review the cardioprotective strategies mediated by caveolins through various pathophysiological pathways. Finally, some possible research directions are proposed to provide future experiments and clinical translation perspectives targeting caveolin based on the investigative evidence.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Jingwen Yong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Haoran Xing
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, 95th Yong An Road, Xuan Wu District, Beijing 100050, PR China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing 100029, PR China.
| |
Collapse
|
6
|
Evaluation of the dystrophin carboxy-terminal domain for micro-dystrophin gene therapy in cardiac and skeletal muscles in the DMD mdx rat model. Gene Ther 2022; 29:520-535. [PMID: 35105949 DOI: 10.1038/s41434-022-00317-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 01/02/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the gene encoding dystrophin. Gene therapy using micro-dystrophin (MD) transgenes and recombinant adeno-associated virus (rAAV) vectors hold great promise. To overcome the limited packaging capacity of rAAV vectors, most MD do not include dystrophin carboxy-terminal (CT) domain. Yet, the CT domain is known to recruit α1- and β1-syntrophins and α-dystrobrevin, a part of the dystrophin-associated protein complex (DAPC), which is a signaling and structural mediator of muscle cells. In this study, we explored the impact of inclusion of the dystrophin CT domain on ΔR4-23/ΔCT MD (MD1), in DMDmdx rats, which allows for relevant evaluations at muscular and cardiac levels. We showed by LC-MS/MS that MD1 expression is sufficient to restore the interactions at a physiological level of most DAPC partners in skeletal and cardiac muscles, and that inclusion of the CT domain increases the recruitment of some DAPC partners at supra-physiological levels. In parallel, we demonstrated that inclusion of the CT domain does not improve MD1 therapeutic efficacy on DMD muscle and cardiac pathologies. Our work highlights new evidences of the therapeutic potential of MD1 and strengthens the relevance of this candidate for gene therapy of DMD.
Collapse
|
7
|
Wang Z, Chan HW, Gambarotta G, Smith NJ, Purdue BW, Pennisi DJ, Porrello ER, O'Brien SL, Reichelt ME, Thomas WG, Paravicini TM. Stimulation of the four isoforms of receptor tyrosine kinase ErbB4, but not ErbB1, confers cardiomyocyte hypertrophy. J Cell Physiol 2021; 236:8160-8170. [PMID: 34170016 DOI: 10.1002/jcp.30487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 11/11/2022]
Abstract
Epidermal growth factor (EGF) receptors (ErbB1-ErbB4) promote cardiac development and growth, although the specific EGF ligands and receptor isoforms involved in growth/repair versus pathology remain undefined. We challenged ventricular cardiomyocytes with EGF-like ligands and observed that selective activation of ErbB4 (the receptor for neuregulin 1 [NRG1]), but not ErbB1 (the receptor for EGF, EGFR), stimulated hypertrophy. This lack of direct ErbB1-mediated hypertrophy occurred despite robust activation of extracellular-regulated kinase 1/2 (ERK) and protein kinase B. Hypertrophic responses to NRG1 were unaffected by the tyrosine kinase inhibitor (AG1478) at concentrations that are selective for ErbB1 over ErbB4. NRG1-induced cardiomyocyte enlargement was suppressed by small interfering RNA (siRNA) knockdown of ErbB4 and ErbB2, whereas ERK phosphorylation was only suppressed by ErbB4 siRNA. Four ErbB4 isoforms exist (JM-a/JM-b and CYT-1/CYT-2), generated by alternative splicing, and their expression declines postnatally and following cardiac hypertrophy. Silencing of all four isoforms in cardiomyocytes, using an ErbB4 siRNA, abrogated NRG1-induced hypertrophic promoter/reporter activity, which was rescued by coexpression of knockdown-resistant versions of the ErbB4 isoforms. Thus, ErbB4 confers cardiomyocyte hypertrophy to NRG1, and all four ErbB4 isoforms possess the capacity to mediate this effect.
Collapse
Affiliation(s)
- Zhen Wang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Hsiu-Wen Chan
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino, Italy
| | - Nicola J Smith
- School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, New South Wales, Australia
| | - Brooke W Purdue
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - David J Pennisi
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Enzo R Porrello
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Shannon L O'Brien
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tamara M Paravicini
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Maternal diet high in linoleic acid alters offspring fatty acids and cardiovascular function in a rat model. Br J Nutr 2021; 127:540-553. [PMID: 33858529 DOI: 10.1017/s0007114521001276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Linoleic acid (LA), an essential n-6 fatty acid (FA), is critical for fetal development. We investigated the effects of maternal high LA (HLA) diet on offspring cardiac development and its relationship to circulating FA and cardiovascular function in adolescent offspring, and the ability of the postnatal diet to reverse any adverse effects. Female Wistar Kyoto rats were fed low LA (LLA; 1·44 % energy from LA) or high LA (HLA; 6·21 % energy from LA) diets for 10 weeks before pregnancy and during gestation/lactation. Offspring, weaned at postnatal day 25, were fed LLA or HLA diets and euthanised at postnatal day 40 (n 6-8). Maternal HLA diet decreased circulating total cholesterol and HDL-cholesterol in females and decreased total plasma n-3 FA in males, while maternal and postnatal HLA diets decreased total plasma n-3 FA in females. α-Linolenic acid (ALA) and EPA were decreased by postnatal but not maternal HLA diets in both sexes. Maternal and postnatal HLA diets increased total plasma n-6 and LA, and a maternal HLA diet increased circulating leptin, in both male and female offspring. Maternal HLA decreased slopes of systolic and diastolic pressure-volume relationship (PVR), and increased cardiac Col1a1, Col3a1, Atp2a1 and Notch1 in males. Maternal and postnatal HLA diets left-shifted the diastolic PVR in female offspring. Coronary reactivity was altered in females, with differential effects on flow repayment after occlusion. Thus, maternal HLA diets impact lipids, FA and cardiac function in offspring, with postnatal diet modifying FA and cardiac function in the female offspring.
Collapse
|
9
|
Terracciano R, Preianò M, Fregola A, Pelaia C, Montalcini T, Savino R. Mapping the SARS-CoV-2-Host Protein-Protein Interactome by Affinity Purification Mass Spectrometry and Proximity-Dependent Biotin Labeling: A Rational and Straightforward Route to Discover Host-Directed Anti-SARS-CoV-2 Therapeutics. Int J Mol Sci 2021; 22:E532. [PMID: 33430309 PMCID: PMC7825748 DOI: 10.3390/ijms22020532] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Protein-protein interactions (PPIs) are the vital engine of cellular machinery. After virus entry in host cells the global organization of the viral life cycle is strongly regulated by the formation of virus-host protein interactions. With the advent of high-throughput -omics platforms, the mirage to obtain a "high resolution" view of virus-host interactions has come true. In fact, the rapidly expanding approaches of mass spectrometry (MS)-based proteomics in the study of PPIs provide efficient tools to identify a significant number of potential drug targets. Generation of PPIs maps by affinity purification-MS and by the more recent proximity labeling-MS may help to uncover cellular processes hijacked and/or altered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), providing promising therapeutic targets. The possibility to further validate putative key targets from high-confidence interactions between viral bait and host protein through follow-up MS-based multi-omics experiments offers an unprecedented opportunity in the drug discovery pipeline. In particular, drug repurposing, making use of already existing approved drugs directly targeting these identified and validated host interactors, might shorten the time and reduce the costs in comparison to the traditional drug discovery process. This route might be promising for finding effective antiviral therapeutic options providing a turning point in the fight against the coronavirus disease-2019 (COVID-19) outbreak.
Collapse
Affiliation(s)
- Rosa Terracciano
- Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy;
| | - Mariaimmacolata Preianò
- Department of Health Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy; (M.P.); (A.F.)
| | - Annalisa Fregola
- Department of Health Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy; (M.P.); (A.F.)
| | - Corrado Pelaia
- Respiratory Medicine Unit, University “Magna Græcia”, 88100 Catanzaro, Italy;
| | - Tiziana Montalcini
- Department of Experimental and Clinical Medicine, University “Magna Græcia”, 88100 Catanzaro, Italy;
| | - Rocco Savino
- Department of Medical and Surgical Sciences, University “Magna Græcia”, 88100 Catanzaro, Italy
| |
Collapse
|
10
|
A Role for Caveolin-3 in the Pathogenesis of Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21228736. [PMID: 33228026 PMCID: PMC7699313 DOI: 10.3390/ijms21228736] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Caveolae are the cholesterol-rich small invaginations of the plasma membrane present in many cell types including adipocytes, endothelial cells, epithelial cells, fibroblasts, smooth muscles, skeletal muscles and cardiac muscles. They serve as specialized platforms for many signaling molecules and regulate important cellular processes like energy metabolism, lipid metabolism, mitochondria homeostasis, and mechano-transduction. Caveolae can be internalized together with associated cargo. The caveolae-dependent endocytic pathway plays a role in the withdrawal of many plasma membrane components that can be sent for degradation or recycled back to the cell surface. Caveolae are formed by oligomerization of caveolin proteins. Caveolin-3 is a muscle-specific isoform, whose malfunction is associated with several diseases including diabetes, cancer, atherosclerosis, and cardiovascular diseases. Mutations in Caveolin-3 are known to cause muscular dystrophies that are collectively called caveolinopathies. Altered expression of Caveolin-3 is also observed in Duchenne’s muscular dystrophy, which is likely a part of the pathological process leading to muscle weakness. This review summarizes the major functions of Caveolin-3 in skeletal muscles and discusses its involvement in the pathology of muscular dystrophies.
Collapse
|
11
|
Russell JS, Griffith TA, Naghipour S, Vider J, Du Toit EF, Patel HH, Peart JN, Headrick JP. Dietary α-Linolenic Acid Counters Cardioprotective Dysfunction in Diabetic Mice: Unconventional PUFA Protection. Nutrients 2020; 12:nu12092679. [PMID: 32887376 PMCID: PMC7551050 DOI: 10.3390/nu12092679] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/12/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022] Open
Abstract
Whether dietary omega-3 (n-3) polyunsaturated fatty acid (PUFA) confers cardiac benefit in cardiometabolic disorders is unclear. We test whether dietary -linolenic acid (ALA) enhances myocardial resistance to ischemia-reperfusion (I-R) and responses to ischemic preconditioning (IPC) in type 2 diabetes (T2D); and involvement of conventional PUFA-dependent mechanisms (caveolins/cavins, kinase signaling, mitochondrial function, and inflammation). Eight-week male C57Bl/6 mice received streptozotocin (75 mg/kg) and 21 weeks high-fat/high-carbohydrate feeding. Half received ALA over six weeks. Responses to I-R/IPC were assessed in perfused hearts. Localization and expression of caveolins/cavins, protein kinase B (AKT), and glycogen synthase kinase-3 β (GSK3β); mitochondrial function; and inflammatory mediators were assessed. ALA reduced circulating leptin, without affecting body weight, glycemic dysfunction, or cholesterol. While I-R tolerance was unaltered, paradoxical injury with IPC was reversed to cardioprotection with ALA. However, post-ischemic apoptosis (nucleosome content) appeared unchanged. Benefit was not associated with shifts in localization or expression of caveolins/cavins, p-AKT, p-GSK3β, or mitochondrial function. Despite mixed inflammatory mediator changes, tumor necrosis factor-a (TNF-a) was markedly reduced. Data collectively reveal a novel impact of ALA on cardioprotective dysfunction in T2D mice, unrelated to caveolins/cavins, mitochondrial, or stress kinase modulation. Although evidence suggests inflammatory involvement, the basis of this "un-conventional" protection remains to be identified.
Collapse
Affiliation(s)
- Jake S. Russell
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Tia A. Griffith
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Saba Naghipour
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Jelena Vider
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Eugene F. Du Toit
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - Hemal H. Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California, San Diego, CA 92093, USA;
| | - Jason N. Peart
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
| | - John P. Headrick
- School of Medical Science, Griffith University Gold Coast, Southport QLD 4217, Australia; (J.S.R.); (T.A.G.); (S.N.); (J.V.); (E.F.D.T.); (J.N.P.)
- Correspondence: ; Tel.: +61-7-5552-8292
| |
Collapse
|
12
|
Lessons from cavin-1 deficiency. Biochem Soc Trans 2020; 48:147-154. [PMID: 31922193 DOI: 10.1042/bst20190380] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 01/19/2023]
Abstract
Caveolae have been implicated in a wide range of critical physiological functions. In the past decade, the dominant role of cavin-1 in caveolae formation has been established, and it has been recognized as another master regulator for caveolae biology. Human patients with cavin-1 mutations develop lipodystrophy and muscular dystrophy and have some major pathological dysfunctions in fat tissue, skeleton muscle, heart, lung and other organs. Cavin-1 deficiency animal models consistently show similar phenotypes. However, the underlying molecular mechanisms remain to be elucidated. Recent studies have suggested many possible pathways, including mechanosensing, stress response, signal transduction, exosome secretion, and potential functions in the nucleus. Many excellent and comprehensive review articles already exist on the topics of caveolae structure formation, caveolins, and their pathophysiological functions. We will focus on recent studies using cavin-1 deficiency models, to summarize the pathophysiological changes in adipose, muscle, and other organs, followed by a summary of mechanistic studies about the roles of cavin-1, which includes caveolae formation, ribosomal RNA transcription, mechanical sensing, stress response, and exosome secretion. Further studies may help to elucidate the exact underlying molecular mechanism to explain the pathological changes observed in cavin-1 deficient human patients and animal models, so potential new therapeutic strategies can be developed.
Collapse
|
13
|
Tian J, Popal MS, Huang R, Zhang M, Zhao X, Zhang M, Song X. Caveolin as a Novel Potential Therapeutic Target in Cardiac and Vascular Diseases: A Mini Review. Aging Dis 2020; 11:378-389. [PMID: 32257548 PMCID: PMC7069461 DOI: 10.14336/ad.2019.09603] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022] Open
Abstract
Caveolin, a structural protein of caveolae, play roles in the regulation of endothelial function, cellular lipid homeostasis, and cardiac function by affecting the activity and biogenesis of nitric oxide, and by modulating signal transduction pathways that mediate inflammatory responses and oxidative stress. In this review, we present the role of caveolin in cardiac and vascular diseases and the relevant signaling pathways involved. Furthermore, we discuss a novel therapeutic perspective comprising crosstalk between caveolin and autophagy.
Collapse
Affiliation(s)
- Jinfan Tian
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mohammad Sharif Popal
- 2 Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - RongChong Huang
- 3 Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100010, China
| | - Min Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xin Zhao
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingduo Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
14
|
Norman R, Fuller W, Calaghan S. Caveolae and the cardiac myocyte. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Morell M, Burgos JI, Gonano LA, Vila Petroff M. AMPK-dependent nitric oxide release provides contractile support during hyperosmotic stress. Basic Res Cardiol 2017; 113:7. [PMID: 29273902 DOI: 10.1007/s00395-017-0665-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/13/2017] [Indexed: 01/11/2023]
Abstract
In different pathological situations, cardiac cells undergo hyperosmotic stress (HS) and cell shrinkage. This change in cellular volume has been associated with contractile dysfunction and cell death. Given that nitric oxide (NO) is a well-recognized modulator of cardiac contractility and cell survival, we evaluated whether HS increases NO production and its impact on the negative inotropic effect observed during this type of stress. Superfusing cardiac myocytes with a hypertonic solution (HS: 440 mOsm) decreased cell volume and increased NO-sensitive DAF-FM fluorescence compared with myocytes superfused with an isotonic solution (IS: 309 mOsm). When cells were exposed to HS in addition to different inhibitors: L-NAME (NO synthase inhibitor), nitroguanidine (nNOS inhibitor), and Wortmannin (eNOS inhibitor) cell shrinkage occurred in the absence of NO release, suggesting that HS activates nNOS and eNOS. Consistently, western blot analysis demonstrated that maintaining cardiac myocytes in HS promotes phosphorylation and thus, activation of nNOS and eNOS compared to myocytes maintained in IS. HS-induced nNOS and eNOS activation and NO production were also prevented by AMPK inhibition with Dorsomorphin (DORSO). In addition, the HS-induced negative inotropic effect was exacerbated in the presence of either L-NAME, DORSO, ODQ (guanylate cyclase inhibitor), or KT5823 (PKG inhibitor), suggesting that NO provides contractile support via a cGMP/PKG-dependent mechanism. Our findings suggest a novel mechanism of AMPK-dependent NO release in cardiac myocytes with putative pathophysiological relevance determined, at least in part, by its capability to reduce the extent of contractile dysfunction associated with hyperosmotic stress.
Collapse
Affiliation(s)
- Malena Morell
- Centro de Investigaciones Cardiovasculares, CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Juan Ignacio Burgos
- Centro de Investigaciones Cardiovasculares, CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Luis Alberto Gonano
- Centro de Investigaciones Cardiovasculares, CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Martin Vila Petroff
- Centro de Investigaciones Cardiovasculares, CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina.
| |
Collapse
|
16
|
Russell J, Du Toit EF, Peart JN, Patel HH, Headrick JP. Myocyte membrane and microdomain modifications in diabetes: determinants of ischemic tolerance and cardioprotection. Cardiovasc Diabetol 2017; 16:155. [PMID: 29202762 PMCID: PMC5716308 DOI: 10.1186/s12933-017-0638-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a ‘wicked triumvirate’: (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia–reperfusion (I–R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.
Collapse
Affiliation(s)
- Jake Russell
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Eugene F Du Toit
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, San Diego, USA
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,School of Medical Science, Griffith University, Southport, QLD, 4217, Australia.
| |
Collapse
|