Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
Cells ; 13(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38667273

RESUMO

Vascular smooth muscle cells (VSMCs), in their contractile and differentiated state, are fundamental for maintaining vascular function. Upon exposure to cholesterol (CHO), VSMCs undergo dedifferentiation, adopting characteristics of foam cells-lipid-laden, macrophage-like cells pivotal in atherosclerotic plaque formation. CHO uptake by VSMCs leads to two primary pathways: ABCA1-mediated efflux or storage in lipid droplets as cholesterol esters (CEs). CE formation, involving the condensation of free CHO and fatty acids, is catalyzed by sterol O-acyltransferase 1 (SOAT1). The necessary fatty acids are synthesized by the lipogenic enzyme fatty acid synthase (FASN), which we found to be upregulated in atherosclerotic human coronary arteries. This observation led us to hypothesize that FASN-mediated fatty acid biosynthesis is crucial in the transformation of VSMCs into foam cells. Our study reveals that CHO treatment upregulates FASN in human aortic SMCs, concurrent with increased expression of CD68 and upregulation of KLF4, markers associated with the foam cell transition. Crucially, downregulation of FASN inhibits the CHO-induced upregulation of CD68 and KLF4 in VSMCs. Additionally, FASN-deficient VSMCs exhibit hindered lipid accumulation and an impaired transition to the foam cell phenotype following CHO exposure, while the addition of the fatty acid palmitate, the main FASN product, exacerbates this transition. FASN-deficient cells also show decreased SOAT1 expression and elevated ABCA1. Notably, similar effects are observed in KLF4-deficient cells. Our findings demonstrate that FASN plays an essential role in the CHO-induced upregulation of KLF4 and the VSMC to foam cell transition and suggest that targeting FASN could be a novel therapeutic strategy to regulate VSMC phenotypic modulation.


Assuntos
Células Espumosas , Fator 4 Semelhante a Kruppel , Músculo Liso Vascular , Animais , Humanos , Aterosclerose/patologia , Aterosclerose/metabolismo , Colesterol/metabolismo , Ácido Graxo Sintases/metabolismo , Ácido Graxo Sintases/genética , Ácidos Graxos/metabolismo , Células Espumosas/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo
2.
Biochem Biophys Res Commun ; 695: 149467, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38211531

RESUMO

Staphylococcus aureus (S. aureus), a Gram-positive bacterium, causes a wide range of infections, and diagnosis at an early stage is challenging. Targeting the maltodextrin transporter has emerged as a promising strategy for imaging bacteria and has been able to image a wide range of bacteria including S. aureus. However, little is known about the maltodextrin transporter in S. aureus, and this prevents new S. aureus specific ligands for the maltodextrin transporter from being developed. In Gram-positive bacteria, including S. aureus, the first step of maltodextrin transport is the binding of the maltodextrin-binding protein malE to maltodextrins. Thus, understanding the binding affinity and characteristics of malE from S. aureus is important to developing efficient maltodextrin-based imaging probes. We evaluated the affinity of malE of S. aureus to maltodextrins of various lengths. MalE of S. aureus (SAmalE) was expressed in E. coli BL21(DE3) and purified by Ni-NTA resin. The affinities of SAmalE to maltodextrins were evaluated with isothermal titration calorimetry. SAmalE has low affinity to maltose but binds to maltotriose and longer maltodextrins up to maltoheptaose with affinities up to Ka = 9.02 ± 0.49 × 105 M-1. SAmalE binding to maltotriose-maltoheptaose was exothermic and fit a single-binding site model. The van't Hoff enthalpy in the binding reaction of SAmalE with maltotriose was 9.9 ± 1.3 kcal/mol, and the highest affinity of SAmalE was observed with maltotetraose with Ka = 9.02 ± 0.49 × 105 M-1. In the plot of ΔH-T*ΔS, the of Enthalpy-Entropy Compensation effect was observed in binding reaction of SAmalE to maltodextrins. Acarbose and maltotetraiol bind with SAmalE indicating that SAmalE is tolerant of modifications on both the reducing and non-reducing ends of maltodextrins. Our results show that unlike ECmalE and similar to the maltodextrin binding protein of Streptococci, SAmalE primarily binds to maltodextrins via hydrogen bonds. This is distinct from the maltodextrin binding protein of Streptococci, SAmalE that binds to maltotetraiol with high affinity. Understanding the binding characteristics and tolerance to maltodextrins modifications by maltodextrin binding proteins will hopefully provide the basis for developing bacterial species-specific maltodextrin-based imaging probes.


Assuntos
Proteínas de Transporte , Staphylococcus aureus , Proteínas de Transporte/metabolismo , Staphylococcus aureus/metabolismo , Escherichia coli/metabolismo , Oligossacarídeos/metabolismo , Proteínas de Bactérias/metabolismo , Polissacarídeos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Calorimetria , Ligação Proteica
3.
Am J Physiol Heart Circ Physiol ; 325(5): H1133-H1143, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37682237

RESUMO

Children with beta-thalassemia (BT) present with an increase in carotid intima-medial thickness, an early sign suggestive of premature atherosclerosis. However, it is unknown if there is a direct relationship between BT and atherosclerotic disease. To evaluate this, wild-type (WT, littermates) and BT (Hbbth3/+) mice, both male and female, were placed on a 3-mo high-fat diet with low-density lipoprotein receptor suppression via overexpression of proprotein convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutation (D377Y). Mechanistically, we hypothesize that heme-mediated oxidative stress creates a proatherogenic environment in BT because BT is a hemolytic anemia that has increased free heme and exhausted hemopexin, heme's endogenous scavenger, in the vasculature. We evaluated the effect of hemopexin (HPX) therapy, mediated via an adeno-associated virus, to the progression of atherosclerosis in BT and a phenylhydrazine-induced model of intravascular hemolysis. In addition, we evaluated the effect of deferiprone (DFP)-mediated iron chelation in the progression of atherosclerosis in BT mice. Aortic en face and aortic root lesion area analysis revealed elevated plaque accumulation in both male and female BT mice compared with WT mice. Hemopexin therapy was able to decrease plaque accumulation in both BT mice and mice on our phenylhydrazine (PHZ)-induced model of hemolysis. DFP decreased atherosclerosis in BT mice but did not provide an additive benefit to HPX therapy. Our data demonstrate for the first time that the underlying pathophysiology of BT leads to accelerated atherosclerosis and shows that heme contributes to atherosclerotic plaque development in BT.NEW & NOTEWORTHY This work definitively shows for the first time that beta-thalassemia leads to accelerated atherosclerosis. We demonstrated that intravascular hemolysis is a prominent feature in beta-thalassemia and the resulting increases in free heme are mechanistically relevant. Adeno-associated virus (AAV)-hemopexin therapy led to decreased free heme and atherosclerotic plaque area in both beta-thalassemia and phenylhydrazine-treated mice. Deferiprone-mediated iron chelation led to deceased plaque accumulation in beta-thalassemia mice but provided no additive benefit to hemopexin therapy.


Assuntos
Doenças da Aorta , Aterosclerose , Placa Aterosclerótica , Talassemia beta , Humanos , Criança , Masculino , Feminino , Camundongos , Animais , Pró-Proteína Convertase 9/genética , Talassemia beta/complicações , Talassemia beta/genética , Hemopexina , Deferiprona , Hemólise , Doenças da Aorta/genética , Doenças da Aorta/patologia , Camundongos Knockout , Aterosclerose/genética , Aterosclerose/patologia , Heme , Fenil-Hidrazinas , Quelantes de Ferro , Camundongos Endogâmicos C57BL
4.
FASEB Bioadv ; 5(5): 199-210, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37151850

RESUMO

The intestinal microbiome has emerged as a potential contributor to the severity of sickle cell disease (SCD). We sought to determine whether SCD mice exhibit intestinal barrier dysfunction, inflammation, and dysbiosis. Using the Townes humanized sickle cell mouse model, we found a 3-fold increase in intestinal permeability as assessed via FITC-dextran (4 kDa) assay in SS (SCD) mice compared to AA (wild type) mice (n = 4, p < 0.05). This was associated with 25 to 50% decreases in claudin-1, 3, and 15 and zonula occludens-1 gene expression (n = 8-10, p < 0.05) in the small intestine. Increased Ly6G staining demonstrated more neutrophils in the SS small intestine (3-fold, n = 5, p < 0.05) associated with increased expression of TNFα, IL-17A, CXCL1, and CD68 (2.5 to 5-fold, n = 7-10, p < 0.05). In addition, we observed 30 to 55% decreases in superoxide dismutase-1, glutathione peroxidase-1, and catalase antioxidant enzyme expression (n = 7-8, p < 0.05) concomitant to an increase in superoxide (2-fold, n = 4, p < 0.05). Importantly, all significant observations of a leaky gut phenotype and inflammation were limited to the small intestine and not observed in the colon. Finally, characterization of the composition of the microbiome within the small intestine revealed dysbiosis in SS mice compared to their AA littermates with 47 phyla to species-level significant alterations in amplicon sequence variants. We conclude that the intestinal barrier is compromised in SCD, associated with decreased gene expression of tight junction proteins, enhanced inflammation, oxidative stress, and gut microbiome dysbiosis, all specific to the small intestine.

5.
Sci Rep ; 13(1): 2404, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36765143

RESUMO

Endothelial dysfunction and inflammatory immune response trigger dedifferentiation of vascular smooth muscle cells (SMCs) from contractile to synthetic phenotype and initiate arterial occlusion. However, the complex vascular remodeling process playing roles in arterial occlusion initiation is largely unknown. We performed bulk sequencing of small and messenger RNAs in a rodent arterial injury model. Bioinformatic data analyses reveal that six miRNAs are overexpressed in injured rat carotids as well as synthetic-type human vascular SMCs. In vitro cell-based assays show that four miRNAs (miR-130b-5p, miR-132-3p, miR-370-3p, and miR-410-3p) distinctly regulate the proliferation of and monocyte adhesion to the vascular SMCs. Individual inhibition of the four selected miRNAs strongly prevents the neointimal hyperplasia in the injured rat carotid arteries. Mechanistically, miR-132-3p and miR-370-3p direct the cell cycle progression, triggering SMC proliferation. Gene ontology analysis of mRNA sequencing data consistently reveal that the miRNA targets include gene clusters that direct proliferation, differentiation, and inflammation. Notably, bone morphogenic protein (BMP)-7 is a prominent target gene of miR-370-3p, and it regulates vascular SMC proliferation in cellular and animal models. Overall, this study first reports that the miR-370-3p/BMP-7 axis determines the vascular SMC phenotype in both rodent and human systems.


Assuntos
MicroRNAs , Músculo Liso Vascular , Animais , Humanos , Ratos , Proteína Morfogenética Óssea 7/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fenótipo
6.
Sci Rep ; 12(1): 8852, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614155

RESUMO

Renewal of the myocardium by preexisting cardiomyocytes is a powerful strategy for restoring the architecture and function of hearts injured by myocardial infarction. To advance this strategy, we show that combining two clinically approved drugs, but neither alone, muscularizes the heart through cardiomyocyte proliferation. Specifically, in adult murine cardiomyocytes, metoprolol, a cardioselective ß1-adrenergic receptor blocker, when given with triiodothyronine (T3, a thyroid hormone) accentuates the ability of T3 to stimulate ERK1/2 phosphorylation and proliferative signaling by inhibiting expression of the nuclear phospho-ERK1/2-specific phosphatase, dual-specificity phosphatase-5. While short-duration metoprolol plus T3 therapy generates new heart muscle in healthy mice, in mice with myocardial infarction-induced left ventricular dysfunction and pathological remodeling, it remuscularizes the heart, restores contractile function and reverses chamber dilatation; outcomes that are enduring. If the beneficial effects of metoprolol plus T3 are replicated in humans, this therapeutic strategy has the potential to definitively address ischemic heart failure.


Assuntos
Infarto do Miocárdio , Disfunção Ventricular Esquerda , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/uso terapêutico , Animais , Metoprolol/farmacologia , Metoprolol/uso terapêutico , Camundongos , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia , Disfunção Ventricular Esquerda/patologia , Remodelação Ventricular
7.
J Biomech Eng ; 144(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35377416

RESUMO

Wall shear stress (WSS) is an important mediator of cardiovascular pathologies and there is a need for its reliable evaluation as a potential prognostic indicator. The purpose of this work was to develop a method that quantifies WSS from two-dimensional (2D) phase contrast magnetic resonance (PCMR) imaging derived flow waveforms, apply this method to PCMR data acquired in the abdominal aorta of healthy volunteers, and to compare PCMR-derived WSS values to values predicted from a computational fluid dynamics (CFD) simulation. The method uses PCMR-derived flow versus time waveforms constrained by the Womersley solution for pulsatile flow in a cylindrical tube. The method was evaluated for sensitivity to input parameters, intrastudy repeatability and was compared with results from a patient-specific CFD simulation. 2D-PCMR data were acquired in the aortas of healthy men (n = 12) and women (n = 15) and time-averaged WSS (TAWSS) was compared. Agreement was observed when comparing TAWSS between CFD and the PCMR flow-based method with a correlation coefficient of 0.88 (CFD: 15.0 ± 1.9 versus MRI: 13.5 ± 2.4 dyn/cm2) though comparison of WSS values between the PCMR-based method and CFD predictions indicate that the PCMR method underestimated instantaneous WSS by 3.7 ± 7.6 dyn/cm2. We found no significant difference in TAWSS magnitude between the sexes; 8.19 ± 2.25 versus 8.07 ± 1.71 dyn/cm2, p = 0.16 for men and women, respectively.


Assuntos
Aorta Abdominal , Modelos Cardiovasculares , Aorta Abdominal/diagnóstico por imagem , Velocidade do Fluxo Sanguíneo , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Estresse Mecânico
8.
Lab Invest ; 102(8): 805-813, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35354915

RESUMO

Sickle cell disease (SCD) is associated with repeated bouts of vascular insufficiency leading to organ dysfunction. Deficits in revascularization following vascular injury are evident in SCD patients and animal models. We aimed to elucidate whether enhancing nitric oxide bioavailability in SCD mice improves outcomes in a model of vascular insufficiency. Townes AA (wild type) and SS (sickle cell) mice were treated with either L-Arginine (5% in drinking water), L-NAME (N(ω)-nitro-L-arginine methyl ester; 1 g/L in drinking water) or NO-generating hydrogel (PA-YK-NO), then subjected to hindlimb ischemia via femoral artery ligation and excision. Perfusion recovery was monitored over 28 days via LASER Doppler perfusion imaging. Consistent with previous findings, perfusion was impaired in SS mice (63 ± 4% of non-ischemic limb perfusion in AA vs 33 ± 3% in SS; day 28; P < 0.001; n = 5-7) and associated with increased necrosis. L-Arginine treatment had no significant effect on perfusion recovery or necrosis (n = 5-7). PA-YK-NO treatment led to worsened perfusion recovery (19 ± 3 vs. 32 ± 3 in vehicle-treated mice; day 7; P < 0.05; n = 4-5), increased necrosis score (P < 0.05, n = 4-5) and a 46% increase in hindlimb peroxynitrite (P = 0.055, n = 4-5). Interestingly, L-NAME worsened outcomes in SS mice with decreased in vivo lectin staining following ischemia (7 ± 2% area in untreated vs 4 ± 2% in treated mice, P < 0.05, n = 5). Our findings demonstrate that L-arginine and direct NO delivery both fail to improve postischemic neovascularization in SCD. Addition of NO to the inflammatory, oxidative environment in SCD may result in further oxidative stress and limit recovery.


Assuntos
Anemia Falciforme , Água Potável , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/metabolismo , Animais , Arginina/metabolismo , Arginina/farmacologia , Disponibilidade Biológica , Água Potável/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia , Camundongos , Músculo Esquelético/metabolismo , NG-Nitroarginina Metil Éster/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Necrose/metabolismo , Neovascularização Fisiológica , Óxido Nítrico/metabolismo , Fluxo Sanguíneo Regional
9.
PLoS One ; 16(12): e0247261, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34928942

RESUMO

POLDIP2 is a multifunctional protein whose roles are only partially understood. Our laboratory previously reported physiological studies performed using a mouse gene trap model, which suffered from three limitations: perinatal lethality in homozygotes, constitutive Poldip2 inactivation and inadvertent downregulation of the adjacent Tmem199 gene. To overcome these limitations, we developed a new conditional floxed Poldip2 model. The first part of the present study shows that our initial floxed mice were affected by an unexpected mutation, which was not readily detected by Southern blotting and traditional PCR. It consisted of a 305 kb duplication around Poldip2 with retention of the wild type allele and could be traced back to the original targeted ES cell clone. We offer simple suggestions to rapidly detect similar accidents, which may affect genome editing using both traditional and CRISPR-based methods. In the second part of the present study, correctly targeted floxed Poldip2 mice were generated and used to produce a new constitutive knockout line by crossing with a Cre deleter. In contrast to the gene trap model, many homozygous knockout mice were viable, in spite of having no POLDIP2 expression. To further characterize the effects of Poldip2 ablation in the vasculature, RNA-seq and RT-qPCR experiments were performed in constitutive knockout arteries. Results show that POLDIP2 inactivation affects multiple cellular processes and provide new opportunities for future in-depth study of its functions.


Assuntos
Sistemas CRISPR-Cas , Marcação de Genes , Proteínas de Membrana/genética , Proteínas Mitocondriais/deficiência , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Nucleares/deficiência , RNA-Seq , Animais , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Proteínas Nucleares/metabolismo
10.
J Am Heart Assoc ; 10(21): e022127, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34689598

RESUMO

Background The growth and remodeling of vascular networks is an important component of the prognosis for patients with peripheral artery disease. One protein that has been previously implicated to play a role in this process is RAGE (receptor for advanced glycation end products). This study sought to determine the cellular source of RAGE in the ischemic hind limb and the role of RAGE signaling in this cell type. Methods and Results Using a hind limb ischemia model of vascular growth, this study found skeletal muscle satellite cells to be a novel major cellular source of RAGE in ischemic tissue by both staining and cellular sorting. Although wild-type satellite cells increased tumor necrosis factor-α and monocyte chemoattractant protein-1 production in response to ischemia in vivo and a RAGE ligand in vitro, satellite cells from RAGE knockout mice lacked the increase in cytokine production both in vivo in response to ischemia and in vitro after stimuli with the RAGE ligand high-mobility group box 1. Furthermore, encapsulated wild-type satellite cells improved perfusion after hind limb ischemia surgery by both perfusion staining and vessel quantification, but RAGE knockout satellite cells provided no improvement over empty capsules. Conclusions Thus, RAGE expression and signaling in satellite cells is crucial for their response to stimuli and angiogenic and arteriogenic functions.


Assuntos
Produtos Finais de Glicação Avançada , Isquemia , Animais , Membro Posterior , Humanos , Isquemia/genética , Ligantes , Camundongos , Receptor para Produtos Finais de Glicação Avançada/genética
11.
Theranostics ; 11(10): 4790-4808, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754028

RESUMO

Rationale: Doxorubicin is a widely used anticancer drug. However, its major side effect, cardiotoxicity, results from cardiomyocyte loss that causes left ventricle (LV) wall thinning, chronic LV dysfunction and heart failure. Cardiomyocyte number expansion by thyroid hormone (T3) during preadolescence is suppressed by the developmental induction of an ERK1/2-specific dual specificity phosphatase 5 (DUSP5). Here, we sought to determine if a brief course of combined DUSP5 suppression plus T3 therapy replaces cardiomyocytes lost due to preexisting doxorubicin injury and reverses heart failure. Methods: We used in vivo-jetPEI to deliver DUSP5 or scrambled siRNA to ~5-week-old C57BL6 mice followed by 5 daily injections of T3 (2 ng/µg body weight). Genetic lineage tracing using Myh6-MerCreMer::Rosa26fs-Confetti mice and direct cardiomyocyte number counting, along with cell cycle inhibition (danusertib), was used to test if this treatment leads to de novo cardiomyocyte generation and improves LV contractile function. Three doses of doxorubicin (20 µg/g) given at 2-weekly intervals, starting at 5-weeks of age in C57BL6 mice, caused severe heart failure, as evident by a decrease in LV ejection fraction. Mice with an ~40 percentage point decrease in LVEF post-doxorubicin injury were randomized to receive either DUSP5 siRNA plus T3, or scrambled siRNA plus vehicle for T3. Age-matched mice without doxorubicin injury served as controls. Results: In uninjured adult mice, transient therapy with DUSP5 siRNA and T3 increases cardiomyocyte numbers, which is required for the associated increase in LV contractile function, since both are blocked by danusertib. In mice with chronic doxorubicin injury, DUSP5 siRNA plus T3 therapy rebuilds LV muscle by increasing cardiomyocyte numbers, which reverses LV dysfunction and prevents progressive chamber dilatation. Conclusion: RNA therapies are showing great potential. Importantly, a GMP compliant in vivo-jetPEI system for delivery of siRNA is already in use in humans, as is T3. Given these considerations, our findings provide a potentially highly translatable strategy for addressing doxorubicin cardiomyopathy, a currently untreatable condition.


Assuntos
Fosfatases de Especificidade Dupla/genética , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Tri-Iodotironina/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Benzamidas/farmacologia , Cardiotoxicidade/etiologia , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Doxorrubicina/toxicidade , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Contração Miocárdica/genética , Miócitos Cardíacos/citologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , RNA Interferente Pequeno , Disfunção Ventricular Esquerda/induzido quimicamente , Função Ventricular Esquerda/genética , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/genética
12.
PLoS One ; 16(3): e0247673, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33647027

RESUMO

Infectious endocarditis is a life-threatening disease, and diagnostics are urgently needed to accurately diagnose this disease especially in the case of prosthetic valve endocarditis. We show here that maltohexaose conjugated to indocyanine green (MH-ICG) can detect Staphylococcus aureus (S. aureus) infection in a rat model of infective endocarditis. The affinity of MH-ICG to S. aureus was determined and had a Km and Vmax of 5.4 µM and 3.0 X 10-6 µmol/minutes/108 CFU, respectively. MH-ICG had no detectable toxicity to mammalian cells at concentrations as high as 100 µM. The in vivo efficiency of MH-ICG in rats was evaluated using a right heart endocarditis model, and the accumulation of MH-ICG in the bacterial vegetations was 2.5 ± 0.2 times higher than that in the control left ventricular wall. The biological half-life of MH-ICG in healthy rats was 14.0 ± 1.3 minutes, and approximately 50% of injected MH-ICG was excreted into the feces after 24 hours. These data demonstrate that MH-ICG was internalized by bacteria with high specificity and that MH-ICG specifically accumulated in bacterial vegetations in a rat model of endocarditis. These results demonstrate the potential efficacy of this agent in the detection of infective endocarditis.


Assuntos
Técnicas de Imagem Cardíaca/métodos , Endocardite Bacteriana/diagnóstico por imagem , Glicoconjugados/química , Verde de Indocianina/química , Oligossacarídeos/química , Infecções Estafilocócicas/diagnóstico por imagem , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Corantes/química , Corantes/farmacocinética , Cricetulus , Modelos Animais de Doenças , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/patologia , Glicoconjugados/farmacocinética , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/microbiologia , Ventrículos do Coração/patologia , Humanos , Verde de Indocianina/farmacocinética , Raios Infravermelhos , Masculino , Oligossacarídeos/farmacocinética , Ratos , Ratos Sprague-Dawley , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidade
13.
Am J Physiol Heart Circ Physiol ; 319(6): H1227-H1233, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32986965

RESUMO

The gut microbiome and intestinal dysfunction have emerged as potential contributors to the development of cardiovascular disease (CVD). Alterations in gut microbiome are well documented in hypertension, atherosclerosis, and heart failure and have been investigated as a therapeutic target. However, a perhaps underappreciated but related role for intestinal barrier function has become evident. Increased intestinal permeability is observed in patients and mouse models of CVD. This increased intestinal permeability can enhance systemic inflammation, alter gut immune function, and has been demonstrated as predictive of adverse cardiovascular outcomes. The goal of this review is to examine the evidence supporting a role for intestinal barrier function in cardiovascular disease and its prospect as a novel therapeutic target. We outline key studies that have investigated intestinal permeability in hypertension, coronary artery disease, atherosclerosis, heart failure, and myocardial infarction. We highlight the central mechanisms involved in the breakdown of barrier function and look at emerging evidence for restored barrier function as a contributor to promising treatment strategies such as short chain fatty acid, probiotic, and renin angiotensin system-targeted therapeutics. Recent studies of more selective targeting of the intestinal barrier to improve disease outcomes are also examined. We suggest that although current data supporting a contribution of intestinal permeability to CVD pathogenesis are largely associative, it appears to be a promising avenue for further investigation. Additional studies of the mechanisms of barrier restoration in CVD and testing of intestinal barrier-targeted compounds will be required to confirm their potential as a new class of CVD therapeutic.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Sistema Cardiovascular/efeitos dos fármacos , Fármacos Gastrointestinais/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/microbiologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/fisiopatologia , Humanos , Intestinos/microbiologia , Intestinos/fisiopatologia , Permeabilidade
14.
Hypertension ; 76(5): 1350-1367, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32981369

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic is associated with significant morbidity and mortality throughout the world, predominantly due to lung and cardiovascular injury. The virus responsible for COVID-19-severe acute respiratory syndrome coronavirus 2-gains entry into host cells via ACE2 (angiotensin-converting enzyme 2). ACE2 is a primary enzyme within the key counter-regulatory pathway of the renin-angiotensin system (RAS), which acts to oppose the actions of Ang (angiotensin) II by generating Ang-(1-7) to reduce inflammation and fibrosis and mitigate end organ damage. As COVID-19 spans multiple organ systems linked to the cardiovascular system, it is imperative to understand clearly how severe acute respiratory syndrome coronavirus 2 may affect the multifaceted RAS. In addition, recognition of the role of ACE2 and the RAS in COVID-19 has renewed interest in its role in the pathophysiology of cardiovascular disease in general. We provide researchers with a framework of best practices in basic and clinical research to interrogate the RAS using appropriate methodology, especially those who are relatively new to the field. This is crucial, as there are many limitations inherent in investigating the RAS in experimental models and in humans. We discuss sound methodological approaches to quantifying enzyme content and activity (ACE, ACE2), peptides (Ang II, Ang-[1-7]), and receptors (types 1 and 2 Ang II receptors, Mas receptor). Our goal is to ensure appropriate research methodology for investigations of the RAS in patients with severe acute respiratory syndrome coronavirus 2 and COVID-19 to ensure optimal rigor and reproducibility and appropriate interpretation of results from these investigations.


Assuntos
Infecções por Coronavirus/epidemiologia , Hipertensão/epidemiologia , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/epidemiologia , Sistema Renina-Angiotensina/fisiologia , Síndrome Respiratória Aguda Grave/metabolismo , Enzima de Conversão de Angiotensina 2 , Determinação da Pressão Arterial/métodos , COVID-19 , China/epidemiologia , Feminino , Humanos , Hipertensão/fisiopatologia , Incidência , Masculino , Pandemias/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Prognóstico , Projetos de Pesquisa , Medição de Risco , Síndrome Respiratória Aguda Grave/epidemiologia
15.
Biochem Biophys Res Commun ; 520(3): 573-579, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31615657

RESUMO

BACKGROUND: Macrophages are ubiquitous in all stages of atherosclerosis, exerting tremendous impact on lesion progression and plaque stability. Because macrophages in atherosclerotic plaques express angiotensin-converting enzyme (ACE), current dogma posits that local myeloid-mediated effects worsen the disease. In contrast, we previously reported that myeloid ACE overexpression augments macrophage resistance to various immune challenges, including tumors, bacterial infection and Alzheimer's plaque deposition. Here, we sought to assess the impact of myeloid ACE on atherosclerosis. METHODS: A mouse model in which ACE is overexpressed in myelomonocytic lineage cells, called ACE10, was generated and sequentially crossed with ApoE-deficient mice to create ACE10/10ApoE-/- (ACE10/ApoE). Control mice were ACEWT/WTApoE-/- (WT/ApoE). Atherosclerosis was induced using an atherogenic diet alone, or in combination with unilateral nephrectomy plus deoxycorticosterone acetate (DOCA) salt for eight weeks. RESULTS: With an atherogenic diet alone or in combination with DOCA, the ACE10/ApoE mice showed significantly less atherosclerotic plaques compared to their WT/ApoE counterparts (p < 0.01). When recipient ApoE-/- mice were reconstituted with ACE10/10 bone marrow, these mice showed significantly reduced lesion areas compared to recipients reconstituted with wild type bone marrow. Furthermore, transfer of ACE-deficient bone marrow had no impact on lesion area. CONCLUSION: Our data indicate that while myeloid ACE may not be required for atherosclerosis, enhanced ACE expression paradoxically reduced disease progression.


Assuntos
Aterosclerose/enzimologia , Aterosclerose/prevenção & controle , Células Mieloides/enzimologia , Peptidil Dipeptidase A/metabolismo , Animais , Aterosclerose/genética , Pressão Sanguínea , Transplante de Medula Óssea , Linhagem da Célula/genética , Colesterol/sangue , Dieta Aterogênica , Modelos Animais de Doenças , Progressão da Doença , Humanos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Células Mieloides/patologia , Peptidil Dipeptidase A/genética , Regulação para Cima
16.
J Cardiovasc Magn Reson ; 21(1): 59, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31522679

RESUMO

BACKGROUND: Displacement Encoding with Stimulated Echoes (DENSE) cardiovascular magnetic resonance (CMR) of the aortic wall offers the potential to improve patient-specific diagnostics and prognostics of diverse aortopathies by quantifying regionally heterogeneous aortic wall strain in vivo. However, before regional mapping of strain can be used to clinically assess aortic pathology, an evaluation of the natural variation of normal regional aortic kinematics is required. METHOD: Aortic spiral cine DENSE CMR was performed at 3 T in 30 healthy adult subjects (range 18 to 65 years) at one or more axial locations that are at high risk for aortic aneurysm or dissection: the infrarenal abdominal aorta (IAA, n = 11), mid-descending thoracic aorta (DTA, n = 17), and/or distal aortic arch (DAA, n = 11). After implementing custom noise-reduction techniques, regional circumferential Green strain of the aortic wall was calculated across 16 sectors around the aortic circumference at each location and normalized by the mean circumferential strain for comparison between individuals. RESULTS: The distribution of normalized circumferential strain (NCS) was heterogeneous for all locations evaluated. Despite large differences in mean strain between subjects, comparisons of NCS revealed consistent patterns of strain distribution for similar groupings of patients by axial location, age, and/or mean displacement angle. NCS at local systole was greatest in the lateral/posterolateral walls in the IAAs (1.47 ± 0.27), medial wall in anteriorly displacing DTAs (1.28 ± 0.20), lateral wall in posteriorly displacing DTAs (1.29 ± 0.29), superior curvature in DAAs < 50 years-old (1.93 ± 0.22), and medial wall in DAAs > 50 years (2.29 ± 0.58). The distribution of strain was strongly influenced by the location of the vertebra and other surrounding structures unique to each location. CONCLUSIONS: Regional in vivo circumferential strain in the adult aorta is unique to each axial location and heterogeneous around its circumference, but can be grouped into consistent patterns defined by basic patient-specific metrics following normalization. The heterogeneous strain distributions unique to each group may be due to local peri-aortic constraints (particularly at the aorto-vertebral interface), heterogeneous material properties, and/or heterogeneous flow patterns. These results must be carefully considered in future studies seeking to clinically interpret or computationally model patient-specific aortic kinematics.


Assuntos
Aorta Abdominal/diagnóstico por imagem , Aorta Torácica/diagnóstico por imagem , Angiografia por Ressonância Magnética , Imagem Cinética por Ressonância Magnética , Modelagem Computacional Específica para o Paciente , Adolescente , Adulto , Idoso , Aorta Abdominal/fisiologia , Aneurisma da Aorta Abdominal/etiologia , Aneurisma da Aorta Abdominal/fisiopatologia , Aneurisma da Aorta Torácica/etiologia , Aneurisma da Aorta Torácica/fisiopatologia , Fenômenos Biomecânicos , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Fluxo Sanguíneo Regional , Reprodutibilidade dos Testes , Fatores de Risco , Estresse Mecânico , Rigidez Vascular , Adulto Jovem
17.
Sci Rep ; 9(1): 9551, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31266969

RESUMO

Critical limb ischemia, the most severe form of peripheral artery disease, leads to extensive damage and alterations to skeletal muscle homeostasis. Although recent research has investigated the tissue-specific responses to ischemia, the role of the muscle stem cell in the regeneration of its niche components within skeletal muscle has been limited. To elucidate the regenerative mechanism of the muscle stem cell in response to ischemic insults, we explored cellular interactions between the vasculature, neural network, and muscle fiber within the muscle stem cell niche. Using a surgical murine hindlimb ischemia model, we first discovered a significant increase in subsynaptic nuclei and remodeling of the neuromuscular junction following ischemia-induced denervation. In addition, ischemic injury causes significant alterations to the myofiber through a muscle stem cell-mediated accumulation of total myonuclei and a concomitant decrease in myonuclear domain size, possibly to enhance the transcriptional and translation output and restore muscle mass. Results also revealed an accumulation of total mitochondrial content per myonucleus in ischemic myofibers to compensate for impaired mitochondrial function and high turnover rate. Taken together, the findings from this study suggest that the muscle stem cell plays a role in motor neuron reinnervation, myonuclear accretion, and mitochondrial biogenesis for skeletal muscle regeneration following ischemic injury.


Assuntos
Extremidades/irrigação sanguínea , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/patologia , Junção Neuromuscular , Animais , Modelos Animais de Doenças , Isquemia/etiologia , Camundongos , Mitocôndrias Musculares/metabolismo , Mioblastos/metabolismo , Regeneração
18.
Circ Res ; 124(4): 607-618, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30763207

RESUMO

Aortic aneurysms are a common vascular disease in Western populations that can involve virtually any portion of the aorta. Abdominal aortic aneurysms are much more common than thoracic aortic aneurysms and combined they account for >25 000 deaths in the United States annually. Although thoracic and abdominal aortic aneurysms share some common characteristics, including the gross anatomic appearance, alterations in extracellular matrix, and loss of smooth muscle cells, they are distinct diseases. In recent years, advances in genetic analysis, robust molecular tools, and increased availability of animal models have greatly enhanced our knowledge of the pathophysiology of aortic aneurysms. This review examines the various proposed cellular mechanisms responsible for aortic aneurysm formation and identifies opportunities for future studies.


Assuntos
Aneurisma Aórtico/metabolismo , Aneurisma Aórtico/epidemiologia , Aneurisma Aórtico/patologia , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Humanos , Estresse Oxidativo , Sistema Renina-Angiotensina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...