Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 319(1): H32-H41, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32412785

RESUMO

Disruption of the normal gut microbiome (dysbiosis) is implicated in the progression and severity of myriad disorders, including hypercholesterolemia and cardiovascular disease. Probiotics attenuate and reverse gut dysbiosis to improve cardiovascular risk factors like hypertension and hypercholesterolemia. Lactobacillus reuteri is a well-studied lactic acid-producing probiotic with known cholesterol-lowering properties and anti-inflammatory effects. In the present study, we hypothesized that L. reuteri delivered to hypercholesterolemic low-density lipoprotein receptor knockout (LDLr KO) mice will reduce cholesterol levels and minimize cardiac injury from an ischemic insult. L. reuteri [1 × 109 or 50 × 106 colony-forming units (CFU)/day] was administered by oral gavage to wild-type mice and LDLr KO for up to 6 wk followed by an ischemia-reperfusion (I/R) protocol. After 4 wk of gavage, total serum cholesterol in wild-type mice receiving saline was 113.5 ± 5.6 mg/dL compared with 113.3 ± 6.8 and 101.9 ± 7.5 mg/dL in mice receiving 1 × 109 or 50 × 106 CFU/day, respectively. Over the same time frame, administration of L. reuteri at 1 × 109 or 50 × 106 CFU/day did not lower total serum cholesterol (283.0 ± 11.1, 263.3 ± 5.0, and 253.1 ± 7.0 mg/dL; saline, 1 × 109 or 50 × 106 CFU/day, respectively) in LDLr KO mice. Despite no impact on total serum cholesterol, L. reuteri administration significantly attenuated cardiac injury following I/R, as evidenced by smaller infarct sizes compared with controls in both wild-type and LDLr KO groups. In conclusion, daily L. reuteri significantly protected against cardiac injury without lowering cholesterol levels, suggesting anti-inflammatory properties of L. reuteri uncoupled from improvements in serum cholesterol.NEW & NOTEWORTHY We demonstrated that daily delivery of Lactobacillus reuteri to wild-type and hypercholesterolemic lipoprotein receptor knockout mice attenuated cardiac injury following ischemia-reperfusion without lowering total serum cholesterol in the short term. In addition, we validated protection against cardiac injury using histology and immunohistochemistry techniques. L. reuteri offers promise as a probiotic to mitigate ischemic cardiac injury.


Assuntos
Colesterol/sangue , Traumatismo por Reperfusão Miocárdica/microbiologia , Probióticos/uso terapêutico , Animais , Microbioma Gastrointestinal , Limosilactobacillus reuteri/patogenicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Probióticos/administração & dosagem , Receptores de LDL/deficiência , Receptores de LDL/genética
2.
Am J Physiol Heart Circ Physiol ; 318(6): H1461-H1473, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32383991

RESUMO

There is a sharp rise in cardiovascular disease (CVD) risk and progression with the onset of menopause. The 4-vinylcyclohexene diepoxide (VCD) model of menopause recapitulates the natural, physiological transition through perimenopause to menopause. We hypothesized that menopausal female mice were more susceptible to CVD than pre- or perimenopausal females. Female mice were treated with VCD or vehicle for 20 consecutive days. Premenopausal, perimenopausal, and menopausal mice were administered angiotensin II (ANG II) or subjected to ischemia-reperfusion (I/R). Menopausal females were more susceptible to pathological ANG II-induced cardiac remodeling and cardiac injury from a myocardial infarction (MI), while perimenopausal, like premenopausal, females remained protected. Specifically, ANG II significantly elevated diastolic (130.9 ± 6.0 vs. 114.7 ± 6.2 mmHg) and systolic (156.9 ± 4.8 vs. 141.7 ± 5.0 mmHg) blood pressure and normalized cardiac mass (15.9 ± 1.0 vs. 7.7 ± 1.5%) to a greater extent in menopausal females compared with controls, whereas perimenopausal females demonstrated a similar elevation of diastolic (93.7 ± 2.9 vs. 100.5 ± 4.1 mmHg) and systolic (155.9 ± 7.3 vs. 152.3 ± 6.5 mmHg) blood pressure and normalized cardiac mass (8.3 ± 2.1 vs. 7.5 ± 1.4%) compared with controls. Similarly, menopausal females demonstrated a threefold increase in fibrosis measured by Picrosirus red staining. Finally, hearts of menopausal females (41 ± 5%) showed larger infarct sizes following I/R injury than perimenopausal (18.0 ± 5.6%) and premenopausal (16.2 ± 3.3, 20.1 ± 4.8%) groups. Using the VCD model of menopause, we provide evidence that menopausal females were more susceptible to pathological cardiac remodeling. We suggest that the VCD model of menopause may be critical to better elucidate cellular and molecular mechanisms underlying the transition to CVD susceptibility in menopausal women.NEW & NOTEWORTHY Before menopause, women are protected against cardiovascular disease (CVD) compared with age-matched men; this protection is gradually lost after menopause. We present the first evidence that demonstrates menopausal females are more susceptible to pathological cardiac remodeling while perimenopausal and cycling females are not. The VCD model permits appropriate examination of how increased susceptibility to the pathological process of cardiac remodeling accelerates from pre- to perimenopause to menopause.


Assuntos
Remodelamento Atrial/fisiologia , Pressão Sanguínea/fisiologia , Doenças Cardiovasculares/fisiopatologia , Cicloexenos , Menopausa/fisiologia , Compostos de Vinila , Angiotensina II , Animais , Doenças Cardiovasculares/induzido quimicamente , Feminino , Camundongos , Modelos Animais
3.
Am J Physiol Heart Circ Physiol ; 311(1): H125-36, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199124

RESUMO

Familial hypertrophic cardiomyopathy (HCM) is a disease of the sarcomere and may lead to hypertrophic, dilated, restrictive, and/or arrhythmogenic cardiomyopathy, congestive heart failure, or sudden cardiac death. We hypothesized that hearts from transgenic HCM mice harboring a mutant myosin heavy chain increase the energetic cost of contraction in a sex-specific manner. To do this, we assessed Ca(2+) sensitivity of tension and crossbridge kinetics in demembranated cardiac trabeculas from male and female wild-type (WT) and HCM hearts at an early time point (2 mo of age). We found a significant effect of sex on Ca(2+) sensitivity such that male, but not female, HCM mice displayed a decrease in Ca(2+) sensitivity compared with WT counterparts. The HCM transgene and sex significantly impacted the rate of force redevelopment by a rapid release-restretch protocol and tension cost by the ATPase-tension relationship. In each of these measures, HCM male trabeculas displayed a gain-of-function when compared with WT counterparts. In addition, cardiac remodeling measured by echocardiography, histology, morphometry, and posttranslational modifications demonstrated sex- and HCM-specific effects. In conclusion, female and male HCM mice display sex dimorphic crossbridge kinetics accompanied by sex- and HCM-dependent cardiac remodeling at the morphometric, histological, and cellular level.


Assuntos
Sinalização do Cálcio , Cardiomiopatia Hipertrófica Familiar/enzimologia , Contração Miocárdica , Miocárdio/enzimologia , Cadeias Pesadas de Miosina/metabolismo , Sarcômeros/enzimologia , Trifosfato de Adenosina/metabolismo , Animais , Cardiomiopatia Hipertrófica Familiar/genética , Cardiomiopatia Hipertrófica Familiar/patologia , Cardiomiopatia Hipertrófica Familiar/fisiopatologia , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Predisposição Genética para Doença , Hidrólise , Cinética , Masculino , Camundongos Transgênicos , Mutação , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Fenótipo , Fosforilação , Caracteres Sexuais , Fatores Sexuais , Remodelação Ventricular
4.
Arch Biochem Biophys ; 601: 32-41, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26971467

RESUMO

Contractile perturbations downstream of Ca(2+) binding to troponin C, the so-called sarcomere-controlled mechanisms, represent the earliest indicators of energy remodeling in the diseased heart [1]. Central to cellular energy "sensing" is the adenosine monophosphate-activated kinase (AMPK) pathway, which is known to directly target myofilament proteins and alter contractility [2-6]. We previously showed that the upstream AMPK kinase, LKB1/MO25/STRAD, impacts myofilament function independently of AMPK [5]. Therefore, we hypothesized that the LKB1 complex associated with myofilament proteins and that alterations in energy signaling modulated targeting or localization of the LKB1 complex to the myofilament. Using an integrated strategy of myofilament mechanics, immunoblot analysis, co-immunoprecipitation, mass spectroscopy, and immunofluorescence, we showed that 1) LKB1 and MO25 associated with myofibrillar proteins, 2) cellular energy stress re-distributed AMPK/LKB1 complex proteins within the sarcomere, and 3) the LKB1 complex localized to the Z-Disk and interacted with cytoskeletal and energy-regulating proteins, including vinculin and ATP Synthase (Complex V). These data represent a novel role for LKB1 complex proteins in myofilament function and myocellular "energy" sensing in the heart.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Miócitos Cardíacos/metabolismo , Miofibrilas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Troponina C/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Cálcio/metabolismo , Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Contração Muscular , Ratos , Ratos Sprague-Dawley , Sarcômeros/metabolismo
5.
Biophys J ; 108(6): 1484-1494, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25809261

RESUMO

The myocardium undergoes extensive metabolic and energetic remodeling during the progression of cardiac disease. Central to remodeling are changes in the adenine nucleotide pool. Fluctuations in these pools can activate AMP-activated protein kinase (AMPK), the central regulator of cellular energetics. Binding of AMP to AMPK not only allosterically activates AMPK but also promotes phosphorylation of AMPK by an upstream kinase complex, LKB1/Mo25/STRAD (liver kinase B 1, mouse protein 25, STE-related adaptor protein). AMPK phosphorylation by the LKB1 complex results in a substantial increase in AMPK activity. Molecular targeting by the LKB1 complex depends on subcellular localization and transcriptional expression. Yet, little is known about the ability of the LKB1 complex to modulate targeting of AMPK after activation. Accordingly, we hypothesized that differing stoichiometric ratios of LKB1 activator complex to AMPK would uniquely impact myofilament function. Demembranated rat cardiac trabeculae were incubated with varying ratios of the LKB1 complex to AMPK or the LKB1 complex alone. After incubation, we measured the Ca(2+) sensitivity of tension, rate constant for tension redevelopment, maximum tension generation, length-dependent activation, cooperativity, and sarcomeric protein phosphorylation status. We found that the Ca(2+) sensitivity of tension and cross-bridge dynamics were dependent on the LKB1 complex/AMPK ratio. We also found that the LKB1 complex desensitizes and suppresses myofilament function independently of AMPK. A phospho-proteomic analysis of myofilament proteins revealed site-specific changes in cardiac Troponin I (cTnI) phosphorylation, as well as a unique distribution of cTnI phosphospecies that were dependent on the LKB1 complex/ AMPK ratio. Fibers treated with the LKB1 complex alone did not alter cTnI phosphorylation or phosphospecies distribution. However, LKB1 complex treatment independent of AMPK increased phosphorylation of myosin-binding protein C. Therefore, we conclude that the LKB1/AMPK signaling axis is able to alter muscle function through multiple mechanisms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Contração Muscular/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Sarcômeros/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio , Coração/fisiologia , Masculino , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos Sprague-Dawley
6.
Physiol Rep ; 11(20): e15838, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37849042

RESUMO

Cardiac ischemic reperfusion injury (IRI) is paradoxically instigated by reestablishing blood-flow to ischemic myocardium typically from a myocardial infarction (MI). Although revascularization following MI remains the standard of care, effective strategies remain limited to prevent or attenuate IRI. We hypothesized that epicardial placement of human placental amnion/chorion (HPAC) grafts will protect against IRI. Using a clinically relevant model of IRI, swine were subjected to 45 min percutaneous ischemia followed with (MI + HPAC, n = 3) or without (MI only, n = 3) HPAC. Cardiac function was assessed by echocardiography, and regional punch biopsies were collected 14 days post-operatively. A deep phenotyping approach was implemented by using histological interrogation and incorporating global proteomics and transcriptomics in nonischemic, ischemic, and border zone biopsies. Our results established HPAC limited the extent of cardiac injury by 50% (11.0 ± 2.0% vs. 22.0 ± 3.0%, p = 0.039) and preserved ejection fraction in HPAC-treated swine (46.8 ± 2.7% vs. 35.8 ± 4.5%, p = 0.014). We present comprehensive transcriptome and proteome profiles of infarct (IZ), border (BZ), and remote (RZ) zone punch biopsies from swine myocardium during the proliferative cardiac repair phase 14 days post-MI. Both HPAC-treated and untreated tissues showed regional dynamic responses, whereas only HPAC-treated IZ revealed active immune and extracellular matrix remodeling. Decreased endoplasmic reticulum (ER)-dependent protein secretion and increased antiapoptotic and anti-inflammatory responses were measured in HPAC-treated biopsies. We provide quantitative evidence HPAC reduced cardiac injury from MI in a preclinical swine model, establishing a potential new therapeutic strategy for IRI. Minimizing the impact of MI remains a central clinical challenge. We present a new strategy to attenuate post-MI cardiac injury using HPAC in a swine model of IRI. Placement of HPAC membrane on the heart following MI minimizes ischemic damage, preserves cardiac function, and promotes anti-inflammatory signaling pathways.


Assuntos
Traumatismos Cardíacos , Infarto do Miocárdio , Gravidez , Suínos , Humanos , Feminino , Animais , Placenta/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Traumatismos Cardíacos/tratamento farmacológico , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Anti-Inflamatórios/uso terapêutico , Modelos Animais de Doenças
7.
Pain ; 163(1): 146-158, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34252907

RESUMO

ABSTRACT: The opioid receptors are important regulators of pain, reward, and addiction. Limited evidence suggests the mu and delta opioid receptors form a heterodimer (MDOR), which may act as a negative feedback brake on opioid-induced analgesia. However, evidence for the MDOR in vivo is indirect and limited, and there are few selective tools available. We recently published the first MDOR-selective antagonist, D24M, allowing us to test the role of the MDOR in mice. We thus cotreated CD-1 mice with D24M and opioids in tail flick, paw incision, and chemotherapy-induced peripheral neuropathy pain models. D24M treatment enhanced oxymorphone antinociception in all models by 54.7% to 628%. This enhancement could not be replicated with the mu and delta selective antagonists CTAP, naltrindole, and naloxonazine, and D24M had a mild transient effect in the rotarod test, suggesting this increase is selective to the MDOR. However, D24M had no effect on morphine or buprenorphine, suggesting that only specific opioids interact with the MDOR. To find a mechanism, we performed phosphoproteomic analysis on brainstems of mice. We found that the kinases Src and CaMKII were repressed by oxymorphone, which was restored by D24M. We were able to confirm the role of Src and CaMKII in D24M-enhanced antinociception using small molecule inhibitors (KN93 and Src-I1). Together, these results provide direct in vivo evidence that the MDOR acts as an opioid negative feedback brake, which occurs through the repression of Src and CaMKII signal transduction. These results further suggest that MDOR antagonism could be a means to improve clinical opioid therapy.


Assuntos
Analgésicos Opioides , Receptores Opioides delta , Analgésicos Opioides/farmacologia , Animais , Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Camundongos , Morfina/farmacologia , Receptores Opioides mu
8.
J Microbiol Methods ; 189: 106302, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34391819

RESUMO

Probiotic strains from the Bifidobacterium or Lactobacillus genera improve health outcomes in models of metabolic and cardiovascular disease. Yet, underlying mechanisms governing these improved health outcomes are rooted in the interaction of gut microbiota, intestinal interface, and probiotic strain. Central to defining the underlying mechanisms governing these improved health outcomes is the development of adaptable and non-invasive tools to study probiotic localization and colonization within the host gut microbiome. The objective of this study was to test labeling and tracking efficacy of Bifidobacterium animalis subspecies lactis 420 (B420) using a common clinical imaging agent, indocyanine green (ICG). ICG was an effective in situ labeling agent visualized in either intact mouse or excised gastrointestinal (GI) tract at different time intervals. Quantitative PCR was used to validate ICG visualization of B420, which also demonstrated that B420 transit time matched normal murine GI motility (~8 hours). Contrary to previous thoughts, B420 did not colonize any region of the GI tract whether following a single bolus or daily administration for up to 10 days. We conclude that ICG may provide a useful tool to visualize and track probiotic species such as B420 without implementing complex molecular and genetic tools. Proof-of-concept studies indicate that B420 did not colonize and establish residency align the murine GI tract.


Assuntos
Bifidobacterium animalis/genética , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Verde de Indocianina/metabolismo , Imagem Óptica/métodos , Animais , Translocação Bacteriana , Bifidobacterium animalis/classificação , Bifidobacterium animalis/isolamento & purificação , Bifidobacterium animalis/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Probióticos , Coloração e Rotulagem
9.
Med Res Arch ; 6(2)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32149188

RESUMO

According to the CDC (2017), more women than men have died from heart disease over the last 20-25 years. On the contrary, premenopausal women are protected against heart and cardiovascular disease (CVD) compared to men. Following menopause, there is sharp rise in CVD mortality and morbidity in women compared to men indicating that women lose protection against CVD during menopause. This loss of CVD protection in women drives the CDC statistics. Life expectance of women has now reached 82 (almost 35 years longer than at the turn of the 20th century). Yet, women typically undergo menopause at 50-60 years of age, which means that women spend over 40% of their life in menopause. Therefore, menopausal women, and associated CVD risk, must be considered as distinct from an aging or senescent woman. Despite longstanding knowledge that premenopausal women are protected from CVD, our fundamental understanding regarding the shift in CVD risk with menopause remains inadequate and impedes our ability to develop sex-specific therapeutic strategies to combat menopausal susceptibility to CVD. This review provides a critical overview of clinical trials attempting to address CVD susceptibility postmenopausal using hormone replacement therapy. Next, we outline key deficiencies in pre-clinical menopause models and introduce an alternative to overcome these deficiencies. Finally, we discuss a novel connection between AMPK and estrogen-dependent pathways that may serve as a potential solution to increased CVD susceptibility in menopausal women.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA