Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Publication year range
1.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35029658

ABSTRACT

Worldwide prevalence of obesity is associated with the increase of lifestyle-related diseases. The accumulation of intermuscular adipose tissue (IMAT) is considered a major problem whereby obesity leads to sarcopenia and metabolic disorders and thus is a promising target for treating these pathological conditions. However, whereas obesity-associated IMAT is suggested to originate from PDGFRα+ mesenchymal progenitors, the processes underlying this adipogenesis remain largely unexplored. Here, we comprehensively investigated intra- and extracellular changes associated with these processes using single-cell RNA sequencing and mass spectrometry. Our single-cell RNA sequencing analysis identified a small PDGFRα+ cell population in obese mice directed strongly toward adipogenesis. Proteomic analysis showed that the appearance of this cell population is accompanied by an increase in galectin-3 in interstitial environments, which was found to activate adipogenic PPARγ signals in PDGFRα+ cells. Moreover, IMAT formation during muscle regeneration was significantly suppressed in galectin-3 knockout mice. Our findings, together with these multi-omics datasets, could unravel microenvironmental networks during muscle regeneration highlighting possible therapeutic targets against IMAT formation in obesity.


Subject(s)
Adipose Tissue/metabolism , Galectin 3/metabolism , Muscle, Skeletal/physiology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Actins/genetics , Actins/metabolism , Adipogenesis , Adipose Tissue/cytology , Animals , Cardiotoxins/pharmacology , Cell Differentiation , Cellular Senescence/genetics , Diet, High-Fat , Female , Galectin 3/deficiency , Galectin 3/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Obesity/metabolism , Obesity/pathology , PPAR gamma/metabolism , Receptor, Platelet-Derived Growth Factor alpha/deficiency , Receptor, Platelet-Derived Growth Factor alpha/genetics , Regeneration , Signal Transduction/genetics
2.
Cell Mol Biol Lett ; 28(1): 27, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37016287

ABSTRACT

BACKGROUND: Innate immune responses play essential roles in skeletal muscle recovery after injury. Programmed cell death protein 1 (PD-1) contributes to skeletal muscle regeneration by promoting macrophage proinflammatory to anti-inflammatory phenotype transition. Interferon (IFN)-γ induces proinflammatory macrophages that appear to hinder myogenesis in vitro. Therefore, we tested the hypothesis that blocking IFN-γ in PD-1 knockout mice may dampen inflammation and promote skeletal muscle regeneration via regulating the macrophage phenotype and neutrophils. METHODS: Anti-IFN-γ antibody was administered in PD-1 knockout mice, and cardiotoxin (CTX) injection was performed to induce acute skeletal muscle injury. Hematoxylin and eosin (HE) staining was used to view morphological changes of injured and regenerated skeletal muscle. Masson's trichrome staining was used to assess the degree of fibrosis. Gene expressions of proinflammatory and anti-inflammatory factors, fibrosis-related factors, and myogenic regulator factors were determined by real-time polymerase chain reaction (PCR). Changes in macrophage phenotype were examined by western blot and real-time PCR. Immunofluorescence was used to detect the accumulation of proinflammatory macrophages, anti-inflammatory macrophages, and neutrophils. RESULTS: IFN-γ blockade in PD-1 knockout mice did not alleviate skeletal muscle damage or improve regeneration following acute cardiotoxin-induced injury. Instead, it exacerbated skeletal muscle inflammation and fibrosis, and impaired regeneration via inhibiting macrophage accumulation, blocking macrophage proinflammatory to anti-inflammatory transition, and enhancing infiltration of neutrophils. CONCLUSION: IFN-γ is crucial for efficient skeletal muscle regeneration in the absence of PD-1.


Subject(s)
Cardiotoxins , Programmed Cell Death 1 Receptor , Animals , Mice , Cardiotoxins/pharmacology , Fibrosis , Inflammation/metabolism , Interferon-gamma/metabolism , Mice, Knockout , Muscle, Skeletal/metabolism , Programmed Cell Death 1 Receptor/metabolism
3.
Pak J Pharm Sci ; 34(3(Special)): 1233-1241, 2021 May.
Article in English | MEDLINE | ID: mdl-34602394

ABSTRACT

To reveal the protective effect of Terminalia chebula Retz (TCR) on cardiotoxicity induced by radix of Aconitum kusnezoffii Reichb (AKR). Control, AKR, AKR-TCR 1:3, AKR-TCR 1:1, AKR-TCR 3:1 and TCR-prepared AKR groups were set up. After treatment, the heart tissues were observed by H&E staining and transmission electron microscope. Serum myoglobin (MB) and troponin (cTn) were detected by ELISA. UPLC-Q Exactive/MS analysis was performed to detect the metabolic difference among the groups. ELISA results showed that the MB and cTn values of AKR group were significantly higher than Control group (P<0.05), while those of the other groups were lower than AKR group. TCR-prepared AKR group had similar MB and cTn contents to the Control group. Histopathological examination also indicated better detoxifying effects in the TCR-prepared AKR and AKR-TCR 1:1 group. The serum metabolomics analysis showed obvious distinction between the AKR and Control groups, while AKR-TCR combination reversed the metabolomics changes induced by AKR. Through multivariate statistical analysis, 9 metabolic markers related to energy, nucleic acid and amino acid metabolism were identified. Conclusively, AKR-induced cardiotoxicity may be related to energy, nucleic acid and amino acid metabolism, and TCR can reduce the cardiotoxicity by regulating the relative metabolism pathways.


Subject(s)
Aconitum , Cardiotoxicity/metabolism , Cardiotoxins/pharmacology , Heart/drug effects , Metabolomics , Myocardium/metabolism , Protective Agents/pharmacology , Terminalia , Amino Acids/drug effects , Amino Acids/metabolism , Animals , Cardiotoxicity/etiology , Energy Metabolism/drug effects , Microscopy, Electron, Transmission , Myocardium/pathology , Myoglobin/blood , Myoglobin/drug effects , Nucleic Acids/drug effects , Nucleic Acids/metabolism , Rats , Troponin/blood , Troponin/drug effects
4.
Expert Rev Proteomics ; 15(11): 873-886, 2018 11.
Article in English | MEDLINE | ID: mdl-30328726

ABSTRACT

INTRODUCTION: Being important representatives of various proteomes, membrane-active cationic peptides (CPs) are attractive objects as lead compounds in the design of new antibacterial, anticancer, antifungal, and antiviral molecules. Numerous CPs are found in insect and snake venoms, where many of them reveal cytolytic properties. Due to advances in omics technologies, the number of such peptides is growing dramatically. Areas covered: To understand structure-function relationships for CPs in a living cell, detailed analysis of their hydrophobic/hydrophilic properties is indispensable. We consider two structural classes of membrane-active CPs: latarcins (Ltc) from spider and cardiotoxins (CTXs) from snake venoms. While the former are void off disulfide bonds and conformationally flexible, the latter are structurally rigid and cross-linked with disulfide bonds. In order to elucidate structure-activity relationships behind their antibacterial, anticancer, and hemolytic effects, the properties of these polypeptides are considered on a side-by-side basis. Expert commentary: An ever-increasing number of venom-derived membrane-active polypeptides require new methods for identification of their functional propensities and sequence-based design of novel pharmacological substances. We address these issues considering a number of the designed peptides, based either on Ltc or CTX sequences. Experimental and computer modeling techniques required for these purposes are delineated.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cardiotoxins/pharmacology , Peptides/chemistry , Peptides/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cardiotoxins/chemistry , Disulfides/chemistry , Drug Design , Hemolytic Agents/chemistry , Hemolytic Agents/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Peptides/metabolism , Spider Venoms/chemistry , Structure-Activity Relationship
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(8): 1904-1911, 2017 08.
Article in English | MEDLINE | ID: mdl-28069395

ABSTRACT

Doxorubicin as anticancer agent can cause dose-dependent cardiotoxicity and heart failure in the long term. Rutin as a polyphenolic flavonoid has been illustrated to protect hearts from diverse cardiovascular diseases. Its function is known to be related to its antioxidant and antiinflammatory activity which may regulate multiple cellular signal pathways. However, the role of rutin on doxorubicin-induced cardiotoxicity has yet to be discovered. In this study, we explored the protective role of rutin on doxorubicin-induced heart failure and elucidated the potential mechanisms of protective effects of rutin against cardiomyocyte death. We analyzed cardiac tissues at the time point of 8weeks after doxorubicin treatment. The results by echocardiography, TUNEL staining, Masson's trichrome staining as well as Western blot analysis revealed that doxorubicin induced remarkable cardiac dysfunction and cardiotoxicity in mice hearts and cardiomyocytes, which were alleviated by rutin treatment. Western blot analysis indicated that the underlying mechanisms included inhibition excessive autophagy and apoptosis mediated by Akt activation. Collectively, our findings suggest that suppression of autophagy and apoptosis by administration of rutin could attenuate doxorubicin-induced cardiotoxicity, which enhances our knowledge to explore new drugs and strategies for combating this devastating side effect induced by doxorubicin. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cardiotoxins/adverse effects , Doxorubicin/adverse effects , Rutin/pharmacology , Animals , Cardiotoxins/pharmacology , Doxorubicin/pharmacology , Male , Mice
6.
Histochem Cell Biol ; 148(1): 49-60, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28293722

ABSTRACT

Tissue damage triggers innate immune response mediated by Toll-like receptor 4 (TLR) that recognizes endogenous host danger molecules associated with cell death and tissue inflammation, although the precise role of TLR-4 signaling in muscle tissue repair is still uncertain. Previously, we observed that TLR-4 exerted a protective effect preventing excessive muscular damage induced by Bothrops jararacussu crude venom. This study aimed to evaluate the involvement of TLR-4 at early stages of muscular tissue remodeling in distinct mouse strains after injection of purified snake venom. Muscular injury was induced by injection of 25 µl (0.05 mg/ml) of cardiotoxin (CTX) from Naja mossambica in the gastrocnemius muscle of C3H/HeN (wild-type); C3H/HeJ mice that express a non-functional TLR-4 receptor, C57BL/6 and Tlr4 -/- (B6 background) mice. Comparing to control, Tlr4 -/- mice presented at early stages (3 DPI) of muscle injury mild inflammation with low MMP-9 activity, scarce macrophage infiltration and premature change to anti-inflammatory phenotype, low TNF-α mRNA levels and reduced myogenin expression, with low regeneration and tissue remodeling. The presence of more Ly6Cneg macrophages in Tlr4 -/- mice at 3 DPI indicates that TLR-4 may influence the differentiation into Ly6Cneg or likely affect proliferation of such cells in the muscle. The present study shows that TLR-4 deficiency and genetic background influence the outcome of muscular tissue repair in aseptic lesions and yet still maintaining some level of signaling in the TLR4-mutant mice.


Subject(s)
Cardiotoxins/pharmacology , Muscle Fibers, Skeletal/drug effects , Toll-Like Receptor 4/deficiency , Animals , Cardiotoxins/administration & dosage , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Toll-Like Receptor 4/metabolism
7.
J Physiol ; 594(18): 5343-62, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27311616

ABSTRACT

KEY POINTS: Mitochondrial-derived vesicle (MDV) formation occurs under baseline conditions and is rapidly upregulated in response to stress-inducing conditions in H9c2 cardiac myoblasts. In mice formation of MDVs occurs readily in the heart under normal healthy conditions while mitophagy is comparatively less prevalent. In response to acute stress induced by doxorubicin, mitochondrial dysfunction develops in the heart, triggering MDV formation and mitophagy. MDV formation is thus active in the cardiac system, where it probably constitutes a baseline housekeeping mechanism and a first line of defence against stress. ABSTRACT: The formation of mitochondrial-derived vesicles (MDVs), a process inherited from bacteria, has emerged as a potentially important mitochondrial quality control (QC) mechanism to selectively deliver damaged material to lysosomes for degradation. However, the existence of this mechanism in various cell types, and its physiological relevance, remains unknown. Our aim was to investigate the dynamics of MDV formation in the cardiac system in vitro and in vivo. Immunofluorescence in cell culture, quantitative transmission electron microscopy and electron tomography in vivo were used to study MDV production in the cardiac system. We show that in cardiac cells MDV production occurs at baseline, is commensurate with the dependence of cells on oxidative metabolism, is more frequent than mitophagy and is up-regulated on the time scale of minutes to hours in response to prototypical mitochondrial stressors (antimycin-A, xanthine/xanthine oxidase). We further show that MDV production is up-regulated together with mitophagy in response to doxorubicin-induced mitochondrial and cardiac dysfunction. Here we provide the first quantitative data demonstrating that MDV formation is a mitochondrial QC operating in the heart.


Subject(s)
Heart/physiology , Mitochondria, Heart/physiology , Animals , Cardiotoxins/pharmacology , Cell Line , DNA, Mitochondrial/genetics , Doxorubicin/pharmacology , Electron Microscope Tomography , Heart/drug effects , Humans , Hydrogen Peroxide/metabolism , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Diseases/genetics , Muscle, Skeletal/diagnostic imaging , Myocardium/ultrastructure , Rats
8.
Hum Mol Genet ; 23(15): 3990-4000, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24659498

ABSTRACT

Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease that causes respiratory and cardiac failure. Inflammation is a key pathological characteristic of dystrophic muscle lesion formation, but its role and regulation in the disease time course has not been sufficiently examined. In the present study, we used IL-10(-/-)/mdx mice lacking both dystrophin and the anti-inflammatory cytokine, interleukin-10 (IL-10), to investigate whether a predisposition to inflammation affects the severity of DMD with advancing age. The IL-10 deficiency caused a profound DMD phenotype in the dystrophic heart such as muscle degeneration and extensive myofiber loss, but the limb muscle and diaphragm morphology of IL-10(-/) (-)/mdx mice was similar to that of mdx mice. Extensive infiltrates of pro-inflammatory M1 macrophages in regeneration of cardiotoxin-injured muscle, altered M1/M2 macrophage phenotype and increased pro-inflammatory cytokines/chemokines production were observed in the diaphragm and heart of IL-10(-/-)/mdx mice. We characterized the IL-10(-/-)/mdx mice as a dystrophic model with chronic inflammation and severe cardiorespiratory dysfunction, as evidenced by decreased percent fractional shortening (%FS) and ejection fraction percent (EF%) on echocardiography, reduced lower tidal volume on whole-body plethysmography. This study suggests that a predisposition to inflammation is an important indicator of DMD disease progression. Therefore, the development of anti-inflammatory strategies may help in slowing down the cardiorespiratory dysfunction on DMD.


Subject(s)
Diaphragm/physiopathology , Dystrophin/genetics , Interleukin-10/genetics , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/physiopathology , Myocardium/pathology , Animals , Cardiotoxins/pharmacology , Diaphragm/metabolism , Dystrophin/deficiency , Female , Gene Expression , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Inflammation/physiopathology , Interleukin-10/deficiency , Lung/metabolism , Lung/physiopathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/complications , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/physiopathology , Myocardium/metabolism , Regeneration , Respiratory Function Tests , Severity of Illness Index , Stroke Volume , Tidal Volume
9.
Blood ; 123(23): 3543-52, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24665135

ABSTRACT

Poor prognosis and limited therapeutic options characterize immunoglobulin light-chain (AL) amyloidosis with major heart involvement. Reliable experimental models are needed to study light-chain (LC)/heart interactions and to explore strategies for prevention of cardiac damage. We have exploited the nematode Caenorhabditis elegans as a novel tool, because its pharynx is evolutionarily related to the vertebrate heart. Our data demonstrate that the pharyngeal pumping of C elegans is significantly and selectively reduced by LCs from AL patients suffering from cardiomyopathy, but not by amyloid LCs with different organ tropism or nonamyloidogenic LCs from multiple myeloma. This functional alteration is dependent on the LC concentration and results in persistent pharyngeal dysfunction and in a significant reduction of the worms' lifespan. These manifestations are paralleled by an increase of mitochondrial reactive oxygen species and can be prevented by treatment with antioxidant agents. In conclusion, these data indicate that this nematode-based assay is a promising surrogate model for investigating the heart-specific toxicity of amyloidogenic LCs and for a rapid screening of new therapeutic strategies.


Subject(s)
Amyloidosis/diagnosis , Caenorhabditis elegans , Heart Diseases/diagnosis , Immunoglobulin Light Chains/immunology , Adult , Aged , Amyloidosis/immunology , Animals , Biological Assay , Cardiotoxins/isolation & purification , Cardiotoxins/pharmacology , Cell Survival/drug effects , Female , Heart Diseases/immunology , Humans , Male , Middle Aged , Multiple Myeloma/immunology , Pharynx/cytology , Pharynx/drug effects , Pharynx/physiology
10.
J Pathol ; 236(2): 229-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25712044

ABSTRACT

Neurological heterotopic ossification (NHO) is the abnormal formation of bone in soft tissues as a consequence of spinal cord or traumatic brain injury. NHO causes pain, ankyloses, vascular and nerve compression and delays rehabilitation in this high-morbidity patient group. The pathological mechanisms leading to NHO remain unknown and consequently there are no therapeutic options to prevent or reduce NHO. Genetically modified mouse models of rare genetic forms of heterotopic ossification (HO) exist, but their relevance to NHO is questionable. Consequently, we developed the first model of spinal cord injury (SCI)-induced NHO in genetically unmodified mice. Formation of NHO, measured by micro-computed tomography, required the combination of both SCI and localized muscular inflammation. Our NHO model faithfully reproduced many clinical features of NHO in SCI patients and both human and mouse NHO tissues contained macrophages. Muscle-derived mesenchymal progenitors underwent osteoblast differentiation in vitro in response to serum from NHO mice without additional exogenous osteogenic stimuli. Substance P was identified as a candidate NHO systemic neuropeptide, as it was significantly elevated in the serum of NHO patients. However, antagonism of substance P receptor in our NHO model only modestly reduced the volume of NHO. In contrast, ablation of phagocytic macrophages with clodronate-loaded liposomes reduced the size of NHO by 90%, supporting the conclusion that NHO is highly dependent on inflammation and phagocytic macrophages in soft tissues. Overall, we have developed the first clinically relevant model of NHO and demonstrated that a combined insult of neurological injury and soft tissue inflammation drives NHO pathophysiology.


Subject(s)
Macrophages/physiology , Myositis/etiology , Ossification, Heterotopic/etiology , Spinal Cord Injuries/complications , Animals , Cardiotoxins/pharmacology , Disease Models, Animal , Female , Humans , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Paraplegia/complications , Stem Cells/physiology
11.
J Immunol ; 193(10): 5149-60, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339660

ABSTRACT

Inflammatory microenvironments play a key role in skeletal muscle regeneration. The infiltration of CD8 T cells into injured muscle has been reported. However, the role of CD8 T cells during skeletal muscle regeneration remains unclear. In this study, we used cardiotoxin-induced mouse skeletal muscle injury/regeneration model to investigate the role of CD8 T cells. Muscle regeneration was impaired and matrix deposit was increased in CD8α-deficient mice compared with wild-type (WT) mice whose CD8 T cells were infiltrated into damaged muscle after cardiotoxin injection. Adoptive transfer of CD8 T cells to CD8α-deficient mice improved muscle regeneration and inhibited matrix remodeling. Compared with WT mice, CD8α deficiency limited the recruitment of Gr1(high) macrophages (MPs) into muscle, resulting in the reduction of satellite cell number. The expression of MCP-1 (MCP-1/CCL2), which regulates the migration of Gr1(high) MPs, was reduced in CD8α-deficient mice compared with WT mice. Coculture CD8 T cells with MPs promoted MCP-1 secretion. The i.m. injection of MCP-1 markedly promoted the recruitment of Gr1(high) MPs and improved muscle regeneration in CD8α-deficient mice. We conclude that CD8 T cells are involved in skeletal muscle regeneration by regulating the secretion of MCP-1 to recruit Gr1(high) MPs, which facilitate myoblast proliferation.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL2/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Receptors, Chemokine/immunology , Animals , CD8 Antigens/genetics , Cardiotoxins/pharmacology , Cell Movement , Cell Proliferation , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Deletion , Gene Expression , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Myoblasts/cytology , Myoblasts/physiology , Receptors, Chemokine/genetics , Regeneration , Signal Transduction
12.
Pediatr Blood Cancer ; 62(3): 502-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25393686

ABSTRACT

BACKGROUND: Surveillance for anthracycline cardiotoxicity in cancer survivors typically utilizes resting M-mode and two-dimensional echocardiography, which are insensitive to detection of subtle myocardial changes. We examined childhood cancer survivors treated with anthracyclines during exercise using various echocardiography techniques to investigate if these tools can better detect subclinical cardiac dysfunction. PROCEDURE: We recruited asymptomatic survivors at least five years post treatment. Echocardiography was performed at rest and at termination of exercise utilizing tissue Doppler techniques and strain rate imaging. RESULTS: Eighty participants were characterized by cardiotoxicity risk status (high [12], moderate [23], low [24], no risk [21]) as defined by the Children's Oncology Group Long Term Follow-Up Guidelines v3.0. The high-risk group had a higher resting heart rate than controls (100 vs. 88 bpm [P for trend = 0.049]). Peak aerobic capacity in all groups was similar. Compared to controls at rest, the high-risk group had evidence of diastolic dysfunction with lower E/A ratios (1.4 vs. 2.0, P = 0.008) and higher septal early diastolic velocities (E/E') of 11.7 versus 9.9 (P = 0.165). With exercise, this difference resolved and myocardial contractile reserve was preserved. CONCLUSIONS: Asymptomatic, pediatric cancer survivors at high-risk for anthracycline cardiotoxicity have some evidence of diastolic filling abnormalities at rest. With exercise, they augment their systolic and diastolic function to achieve normal maximal aerobic capacity suggesting they are able to compensate for mild cardiac dysfunction in the early years after exposure. Additionally, findings suggest that routine exercise echocardiography may not be a useful surveillance tool to assess anthracycline cardiotoxicity.


Subject(s)
Anthracyclines/adverse effects , Cardiotoxins/adverse effects , Echocardiography, Stress , Exercise , Heart Diseases , Heart Rate , Neoplasms , Survivors , Adolescent , Adult , Anthracyclines/pharmacology , Cardiotoxins/pharmacology , Child , Female , Heart Diseases/chemically induced , Heart Diseases/diagnostic imaging , Heart Diseases/physiopathology , Humans , Male , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/physiopathology
13.
Xenobiotica ; 45(11): 1024-9, 2015.
Article in English | MEDLINE | ID: mdl-25970816

ABSTRACT

1. The clinical use of doxorubicin, an effective anticancer drug, is severely hampered by its cardiotoxicity. Berberine, a botanical alkaloid, has been reported to possess cardioprotective and antitumor effects. In this study, we investigated the cardioprotective effect of berberine on doxorubicin-induced cardiotoxicity and the effect of berberine on the metabolism of doxorubicin. 2. Adult male Sprague-Dawley rats were administered doxorubicin in the presence or absence of berberine for 2 weeks. Administration of berberine effectively prevented doxorubicin-induced body weight reduction and mortality in rats. 3. Berberine reduced the activity of myocardial enzymes, including aspartate aminotransferase (AST), creatine kinase (CK), CK isoenzyme (CK-MB) and lactate dehydrogenase (LDH). Echocardiographic examination further demonstrated that berberine effectively ameliorated cardiac dysfunction induced by doxorubicin. 4. Berberine inhibited the metabolism of doxorubicin in the cytoplasm of rat heart and reduced the accumulation of doxorubicinol (a secondary alcohol metabolite of doxorubicin) in heart. 5. These data showed that berberine alleviated the doxorubicin-induced cardiotoxicity in rats via inhibition of the metabolism of doxorubicin and reduced accumulation of doxorubicinol selectively in hearts.


Subject(s)
Berberine/pharmacology , Cardiomyopathies/congenital , Cardiomyopathies/drug therapy , Cardiotoxins , Doxorubicin , Animals , Cardiomyopathies/metabolism , Cardiotoxins/adverse effects , Cardiotoxins/pharmacokinetics , Cardiotoxins/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Male , Rats , Rats, Sprague-Dawley
14.
Mol Pharmacol ; 86(4): 438-49, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106424

ABSTRACT

The use of anthracycline chemotherapeutic drugs is restricted owing to potentially fatal cardiotoxic side effects. It has been hypothesized that anthracycline metabolites have a primary role in this cardiac dysfunction; however, information on the molecular interactions of these compounds in the heart is scarce. Here we provide novel evidence that doxorubicin and its metabolite, doxorubicinol, bind to the cardiac ryanodine receptor (RyR2) and to the sarco/endoplasmic reticulum Ca(2+) ATPase (SERCA2A) and deleteriously alter their activity. Both drugs (0.01 µM-2.5 µM) activated single RyR2 channels, and this was reversed by drug washout. Both drugs caused a secondary inhibition of RyR2 activity that was not reversed by drug washout. Preincubation with the reducing agent dithiothreitol (DTT, 1 mM) prevented drug-induced inhibition of channel activity. Doxorubicin and doxorubicinol reduced the abundance of thiol groups on RyR2, further indicating that oxidation reactions may be involved in the actions of the compounds. Ca(2+) uptake into sarcoplasmic reticulum vesicles by SERCA2A was inhibited by doxorubicinol, but not doxorubicin. Unexpectedly, in the presence of DTT, doxorubicinol enhanced the rate of Ca(2+) uptake by SERCA2A. Together the evidence provided here shows that doxorubicin and doxorubicinol interact with RyR2 and SERCA2A in similar ways, but that the metabolite acts with greater efficacy than the parent compound. Both compounds modify RyR2 and SERCA2A activity by binding to the proteins and also act via thiol oxidation to disrupt SR Ca(2+) handling. These actions would have severe consequences on cardiomyocyte function and contribute to clinical symptoms of acute anthracycline cardiotoxicity.


Subject(s)
Calcium Signaling/drug effects , Doxorubicin/analogs & derivatives , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Calcium/metabolism , Cardiotoxins/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Lipid Bilayers/metabolism , Protein Binding , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sheep
15.
J Biol Chem ; 288(3): 1489-99, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23184935

ABSTRACT

Inflammation and microenvironment play a crucial role in muscle regeneration. IL (interleukin)-6, as a multifunctional cytokine is involved in the processes. However, the causative effect of IL-6 in muscle regeneration remains unclear. In a mouse model of cardiotoxin-induced muscle injury/regeneration, infiltrated monocytes/macrophages produce a high level of IL-6 started on 1 day (24 h) after injury. In IL-6 knock-out (-/-) mice, the muscle regeneration procedure was impaired along with decreased myogenic determination factor (MyoD) and myogenin mRNA level and increased interstitial fibrosis. The IL-6(-/-) mice exhibited less macrophage infiltration, lower inflammatory cytokine (IL-1ß, inducible NO synthase, Transforming growth factor (TGF)-ß1, and IL-10) and chemokine (CCL2, CCL3, and CCL5) expression, and inhibited myoblast proliferation. In vitro, IL-6 deficiency or Signal Transducer and Activator of Transcription 3 (STAT3) knockdown in activated macrophage attenuated the expression of CCL2, CCL3, but not CCL5, which resulted in less macrophage migration. Moreover, inflammatory macrophages promoted myoblast proliferation in an IL-6-dependent manner. Finally, adoptive transfer IL-6(+/+) BM cells into IL-6(-/-) mice rescued the impaired regeneration with improved MyoD and myogenin expression. Taken together, IL-6 expression and the activated STAT3 signaling pathway in monocytes/macrophages is a critical mediator of macrophage migration and myoblast proliferation during muscle regeneration.


Subject(s)
Interleukin-6/genetics , Macrophages/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Regeneration/genetics , STAT3 Transcription Factor/genetics , Animals , Cardiotoxins/pharmacology , Cell Movement , Cell Proliferation , Gene Expression Regulation , Interleukin-6/metabolism , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myogenin/genetics , Myogenin/metabolism , Primary Cell Culture , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
16.
Anal Chem ; 86(4): 2213-20, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24456565

ABSTRACT

Given the high mortality in patients with cardiovascular diseases and the life-threatening consequences of drugs with unforeseen adverse effects on hearts, a critical evaluation of the pharmacological response of cardiovascular function on model animals is important especially in the early stages of drug development. We report a proof-of-principle study to demonstrate the utility of zebrafish as an analytical platform to predict the cardiac response of new drugs or chemicals on human beings. With pseudodynamic 3D imaging, we derive individual parameters that are central to the cardiac function of zebrafish, including the ventricular stroke volume, ejection fraction, cardiac output, heart rate, diastolic filling function, and ventricular mass. We evaluate both inotropic and chronotropic responses of the heart of zebrafish treated with drugs that are commonly prescribed and possess varied known cardiac activities. We reveal deranged cardiac function of a zebrafish model of cardiomyopathy induced with a cardiotoxic drug. The cardiac function of zebrafish exhibits a pharmacological response similar to that of human beings. We compare also cardiac parameters obtained in this work with those derived with conventional 2D approximation and show that the latter tends to overestimate the cardiac parameters and produces results of greater variation. In view of the growing interest of using zebrafish in both fundamental and translational biomedical research, we envisage that our approach should benefit not only contemporary pharmaceutical development but also exploratory research such as gene, stem cell, or regenerative therapies targeting congenital or acquired heart diseases.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotoxins/pharmacology , Heart/drug effects , Imaging, Three-Dimensional/methods , Animals , Animals, Genetically Modified , Drug Evaluation, Preclinical/methods , Microscopy, Confocal/methods , Zebrafish
17.
Development ; 138(17): 3639-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21828092

ABSTRACT

Skeletal muscle tissue provides mechanical force for locomotion of all vertebrate animals. It is prone to damage from acute physical trauma and physiological stress. To cope with this, it possesses a tremendous capacity for rapid and effective repair that is widely held to be accomplished by the satellite cells lying between the muscle fiber plasmalemma and the basement membrane. Cell transplantation and lineage-tracing studies have demonstrated that Pax7-expressing (Pax7(+)) satellite cells can repair damaged muscle tissue repeatedly after several bouts of acute injury. These findings provided evidence that Pax7(+) cells are muscle stem cells. However, stem cells from a variety of other origins are also reported to contribute to myofibers upon engraftment into muscles, questioning whether satellite cells are the only stem cell source for muscle regeneration. Here, we have engineered genetic ablation of Pax7(+) cells to test whether there is any significant contribution to muscle regeneration after acute injury from cells other than this source. We find that such elimination of Pax7(+) cells completely blocks regenerative myogenesis either following injury to the tibialis anterior (TA) muscle or after transplantation of extensor digitorum longus (EDL) muscles into nude mice. As Pax7 is specifically expressed in satellite cells, we conclude that they are essential for acute injury-induced muscle regeneration. It remains to be established whether there is any significant role for stem cells of other origins. The implications of our results for muscle stem cell-based therapy are discussed.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Animals , Bungarotoxins/pharmacology , Cardiotoxins/pharmacology , Cell Proliferation/drug effects , Diphtheria Toxin/pharmacology , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Mutant Strains , Mice, Nude , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , PAX7 Transcription Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/physiology , Tamoxifen/pharmacology
18.
Mol Ther ; 21(8): 1551-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23752313

ABSTRACT

In the context of future adeno-associated viral (AAV)-based clinical trials for Duchenne myopathy, AAV genome fate in dystrophic muscles is of importance considering the viral capsid immunogenicity that prohibits recurring treatments. We showed that AAV genomes encoding non-therapeutic U7 were lost from mdx dystrophic muscles within 3 weeks after intramuscular injection. In contrast, AAV genomes encoding U7ex23 restoring expression of a slightly shortened dystrophin were maintained endorsing that the arrest of the dystrophic process is crucial for maintaining viral genomes in transduced fibers. Indeed, muscles treated with low doses of AAV-U7ex23, resulting in sub-optimal exon skipping, displayed much lower titers of viral genomes, showing that sub-optimal dystrophin restoration does not prevent AAV genome loss. We also followed therapeutic viral genomes in severe dystrophic dKO mice over time after systemic treatment with scAAV9-U7ex23. Dystrophin restoration decreased significantly between 3 and 12 months in various skeletal muscles, which was correlated with important viral genome loss, except in the heart. Altogether, these data show that the success of future AAV-U7 therapy for Duchenne patients would require optimal doses of AAV-U7 to induce substantial levels of dystrophin to stabilize the treated fibers and maintain the long lasting effect of the treatment.


Subject(s)
Alternative Splicing , Dependovirus/genetics , Genetic Vectors/genetics , Genome, Viral , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , RNA, Small Nuclear/genetics , Animals , Cardiotoxins/pharmacology , Dependovirus/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Exons , Gene Expression , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/metabolism , Humans , Injections, Intramuscular , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy
19.
Birth Defects Res B Dev Reprod Toxicol ; 101(5): 371-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25250843

ABSTRACT

The use of antidepressant drug bupropion hydrochloride (BPN) during pregnancy results in increased cardiovascular anomalies. In this study, BPN developmental cardiotoxic effects in in vitro system were evaluated using chick cardiomyocyte micromass (MM) culture system and mouse embryonic stem cell derived cardiomyocyte (ESDC) system. In MM system, the cardiomyocyte contractile activity significantly decreased only at BPN 200 µM, while in ESDC system BPN concentration above 75 µM resulted in decreased contractile activity. The increase in drug concentration also affected the cardiomyocyte viability and total cellular protein content in both systems, but in ESDC system the cell viability failed to attain significant difference. The drug failed to induce reactive oxygen species production in both systems, but has affected the cardiac connexin43 expression especially in MM system. We observed that BPN showed developmental cardiotoxic effects irrespective of the stage of cardiac development in both in vitro systems.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Bupropion/pharmacology , Cardiotoxins/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Animals , Bupropion/adverse effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chickens , Connexin 43 , Embryonic Stem Cells/cytology , Female , Heart/embryology , Mice , Pregnancy , Reactive Oxygen Species/metabolism , Stem Cells
20.
Pediatr Cardiol ; 35(1): 82-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23821296

ABSTRACT

We aimed to assess early-onset chronic progressive cardiotoxicity in the left and right ventricles with increasing cumulative anthracycline doses. We evaluated 72 patients within the first year after doxorubicin and/or daunorubicin treatment (median 1.3 months; range 0.3-11.5) and 31 healthy controls. Pretreatment and posttreatment QT interval analyzes were performed in 27 newly diagnosed patients. The echocardiographic data of all examinations of 72 patients were classified into three groups according to instant cumulative anthracycline doses: treatment group (TG)-I (≤120 mg/m(2); n = 26), TG-II (120-240 mg/m(2); n = 39), and TG-III (≥240 mg/m(2); n = 40). Diastolic and systolic parameters were analyzed by conventional echocardiography and tissue Doppler imaging (TDI) and compared with those of healthy controls. The mean age for patients and controls was 8.2 ± 4.5 and 9.6 ± 4.2 years, respectively (p > 0.05). QTc dispersion significantly increased after anthracycline treatment (p = 0.02). TDI showed decreased E' velocity (p < 0.001) and E'/A' ratio (p < 0.001) at lateral tricuspid annulus segment in TG-I, and these findings continued in TG-II and -III. In addition, S' velocity decreased in TG-I, -II, and -III at lateral mitral annulus (10.5 ± 2.6 cm/s, p < 0.05; 9.9 ± 2.2 cm/s, p < 0.001; and 10.1 ± 2.3 cm/s, p < 0.01, respectively). However, decrease in left-ventricular ejection fraction was statistically significant in TG-II and -III (p < 0.001). Although myocardial performance index was significantly increased in all treatment groups in both segments, it was primarily due to significant increases in isovolumic relaxation time at the lateral tricuspid annulus and isovolumic contraction time at the lateral mitral annulus. Abnormalities in diastolic function in right ventricle and systolic function in the left ventricle were observed even with a cumulative anthracycline dose <120 mg/m(2) by TDI. In addition, anthracycline treatment led to an increase in QTc dispersion.


Subject(s)
Anthracyclines/pharmacology , Heart Ventricles , Ventricular Dysfunction , Antibiotics, Antineoplastic/pharmacology , Cardiotoxins/pharmacology , Child , Child, Preschool , Chronic Disease , Dose-Response Relationship, Drug , Echocardiography/methods , Electrocardiography/methods , Female , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Humans , Male , Stroke Volume/drug effects , Time Factors , Turkey , Ventricular Dysfunction/chemically induced , Ventricular Dysfunction/diagnosis , Ventricular Dysfunction/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL