Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Circ Res ; 130(10): 1550-1564, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35430873

RESUMO

BACKGROUND: Renal T cells contribute importantly to hypertension, but the underlying mechanism is incompletely understood. We reported that CD8Ts directly stimulate distal convoluted tubule cells (DCTs) to increase NCC (sodium chloride co-transporter) expression and salt reabsorption. However, the mechanistic basis of this pathogenic pathway that promotes hypertension remains to be elucidated. METHODS: We used mouse models of DOCA+salt (DOCA) treatment and adoptive transfer of CD8+ T cells (CD8T) from hypertensive animals to normotensive animals in in vivo studies. Co-culture of mouse DCTs and CD8Ts was used as in vitro model to test the effect of CD8T activation in promoting NCC-mediated sodium retention and to identify critical molecular players contributing to the CD8T-DCT interaction. Interferon (IFNγ)-KO mice and mice receiving renal tubule-specific knockdown of PDL1 were used to verify in vitro findings. Blood pressure was continuously monitored via radio-biotelemetry, and kidney samples were saved at experimental end points for analysis. RESULTS: We identified critical molecular players and demonstrated their roles in augmenting the CD8T-DCT interaction leading to salt-sensitive hypertension. We found that activated CD8Ts exhibit enhanced interaction with DCTs via IFN-γ-induced upregulation of MHC-I and PDL1 in DCTs, thereby stimulating higher expression of NCC in DCTs to cause excessive salt retention and progressive elevation of blood pressure. Eliminating IFN-γ or renal tubule-specific knockdown of PDL1 prevented T cell homing into the kidney, thereby attenuating hypertension in 2 different mouse models. CONCLUSIONS: Our results identified the role of activated CD8Ts in contributing to increased sodium retention in DCTS through the IFNγ-PDL1 pathway. These findings provide a new mechanism for T cell involvement in the pathogenesis of hypertension and reveal novel therapeutic targets.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão , Animais , Linfócitos T CD8-Positivos/metabolismo , Acetato de Desoxicorticosterona/metabolismo , Acetato de Desoxicorticosterona/farmacologia , Modelos Animais de Doenças , Hipertensão/metabolismo , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/patologia , Camundongos , Sódio/metabolismo , Simportadores de Cloreto de Sódio/metabolismo , Cloreto de Sódio na Dieta
2.
Adv Physiol Educ ; 46(4): 728-734, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36264925

RESUMO

Engaging preclinical medical students in the curriculum is challenging. To address this challenge, the investigators developed and implemented self-paced polling with recorded lectures, in which students answered audience response questions at their own pace. In 2021, we retrospectively assigned second-year medical students (N = 165) as Active or Inactive based on their answered polling questions. We subdivided the Active group into two groups, a Live group who predominantly responded to polling in live classes and a Self-paced group who predominantly used polling with recorded lectures. Outcomes were academic performance on customized National Board of Medical Examiners (NBME) examinations and engagement. Compared with the Inactive group, the Self-paced group performed better on the customized NBME examination after extensive self-paced polling. Students answered a significantly larger proportion of questions correctly in self-paced polling compared with live polling. Students who used self-paced polling reported a positive experience and indicated they had emotional, behavioral, or cognitive engagement with the curriculum. This study introduces self-paced polling with recorded lectures, which medical educators can potentially use to enhance student engagement and academic performance.NEW & NOTEWORTHY More medical students utilize recorded lectures than live lectures. Self-paced polling questions allow students to participate while watching recorded lectures. Second-year medical students performed significantly better on examination after actively using the self-paced polling compared with inactive students. They also reported emotional, behavioral, and cognitive engagement with the course material while using the self-paced polling.


Assuntos
Desempenho Acadêmico , Estudantes de Medicina , Humanos , Estudantes de Medicina/psicologia , Estudos Retrospectivos , Inquéritos e Questionários , Currículo , Avaliação Educacional
3.
Adv Physiol Educ ; 46(1): 35-40, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34709944

RESUMO

Retrieval practice improves long-term retention. Use of interactive retrieval practice in large group, in-person and online live classes, in combination with outside resources, is unreported for medical physiology classes. The primary study purpose was to compare student cohorts' performance with or without retrieval practice in renal physiology classes, relative to the national average on customized national examinations in renal physiology, nonphysiology, and all questions. The secondary purpose was to examine the students' educational experience. For the primary purpose, we used a nonequivalent group, posttest-only design. For the secondary purpose, we used cross-sectional and qualitative designs. We analyzed examination results of 684 students in four academic years. For renal physiology questions, students performed significantly better in years with retrieval practice compared with years without it (P < 0.001). There was no change in nonphysiology scores over the four years. Performance in all questions, too, significantly improved (P < 0.001). A large majority (86%) of students indicated retrieval practice helped them learn renal physiology. Student ratings of quality in online classes, which featured interactive retrieval practice, were higher than that of in-person classes (P < 0.001). Qualitative analysis revealed students found interactive retrieval practice, scaffolding, outside resources, and the instructor's teaching style helpful. Educators in medical physiology classes can use our findings to implement interactive retrieval practice.


Assuntos
Estudantes de Medicina , Médicos Legistas , Estudos Transversais , Avaliação Educacional , Humanos , Aprendizagem
4.
J Pharmacol Exp Ther ; 376(1): 127-135, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33100271

RESUMO

The practice of prescribing ß-blockers to lower blood pressure and mitigate perioperative cardiovascular events has been questioned because of reports of an increased risk of stroke. The benefit of ß-blocker therapy primarily relies on preventing activation of cardiac ß1-adrenergic receptors (ARs). However, we reported that ß1ARs also mediate vasodilator responses of rat cerebral arteries (CAs), implying that ß-blockers may impair cerebral blood flow under some conditions. Here, we defined the impact of metoprolol (MET), a widely prescribed ß1AR-selective antagonist, on adrenergic-elicited diameter responses of rat CAs ex vivo and in vivo. MET (1-10 µmol/l) prevented ß1AR-mediated increases in diameter elicited by dobutamine in cannulated rat CAs. The ß1AR-mediated dilation elicited by the endogenous adrenergic agonist norepinephrine (NE) was reversed to a sustained constriction by MET. Acute oral administration of MET (30 mg/kg) to rats in doses that attenuated resting heart rate and dobutamine-induced tachycardia also blunted ß1AR-mediated dilation of CAs. In the same animals, NE-induced dilation of CAs was reversed to sustained constriction. Administration of MET for 2 weeks in drinking water (2 mg/ml) or subcutaneously (15 mg/kg per day) also resulted in NE-induced constriction of CAs in vivo. Thus, doses of MET that protect the heart from adrenergic stimulation also prevent ß1AR-mediated dilation of CAs and favor anomalous adrenergic constriction. Our findings raise the possibility that the increased risk of ischemic stroke in patients on ß-blockers relates in part to adrenergic dysregulation of cerebrovascular tone. SIGNIFICANCE STATEMENT: ß-Blocker therapy using second-generation, cardioselective ß-blockers is associated with an increased risk of stroke, but the responsible mechanisms are unclear. Here, we report that either acute or chronic systemic administration of a cardioselective ß-blocker, metoprolol, mitigates adrenergic stimulation of the heart as an intended beneficial action. However, metoprolol concomitantly eliminates vasodilator responses to adrenergic stimuli of rat cerebral arteries in vivo as a potential cause of dysregulated cerebral blood flow predisposing to ischemic stroke.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Cardiotônicos/farmacologia , Artérias Cerebrais/efeitos dos fármacos , Metoprolol/farmacologia , Receptores Adrenérgicos beta 1/metabolismo , Vasodilatação , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/administração & dosagem , Antagonistas de Receptores Adrenérgicos beta 1/efeitos adversos , Animais , Cardiotônicos/administração & dosagem , Cardiotônicos/efeitos adversos , Artérias Cerebrais/fisiologia , Dobutamina/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Masculino , Metoprolol/administração & dosagem , Metoprolol/efeitos adversos , Norepinefrina/farmacologia , Ratos , Ratos Sprague-Dawley
5.
BMC Med Educ ; 20(1): 87, 2020 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209076

RESUMO

BACKGROUND: Strong learner-teacher relationships are associated with more successful learning outcomes. With shortened modular curricula and increased availability of online resources, fostering faculty interaction with preclinical medical students has become more challenging. We sought to enhance learner-teacher relationships by engaging in discussion with preclinical medical students in their own online space. METHODS: We utilized a closed Facebook discussion group, where faculty and students voluntarily joined in informal discussions and shared announcements related to their courses. The closed discussion group allowed only participating students and faculty to see others' posts within the group. This provided a platform to freely interact within the confines of the group while maintaining privacy for the personal Facebook accounts of both faculty and students. We utilized the discussion group through three separate organ system-based modules for 14 weeks. Afterward, students were asked to complete an anonymous, voluntary online survey about their experience. RESULTS: 94.1% (160/170) of enrolled second-year medical students joined the voluntary FB discussion group. There were 214 posts, 628 comments, and 4166 reactions in this discussion group during the three modules. Of the students in the group, 74.4% (119/160) responded to the online survey. Overall, students strongly agreed that the Facebook discussion group fostered better rapport with faculty, helped content learning, and improved emotional well-being. Also, they felt more comfortable seeking academic help after using the discussion group. They reported a slight preference for Facebook over email as a medium for asking questions, but no preference for either as a medium for distributing announcements. Students overwhelmingly recommended that the discussion group should be continued in future years. CONCLUSION: The Facebook discussion group was a free, efficient, and effective method of cultivating the learner-teacher relationship with the preclinical medical students, resulting in reported enhancement of learning and morale.


Assuntos
Comunicação , Docentes , Mídias Sociais , Estudantes de Medicina , Educação Médica , Humanos , Estudos Retrospectivos , Inquéritos e Questionários
6.
J Pharmacol Exp Ther ; 371(2): 278-289, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31439806

RESUMO

Doxorubicin is a risk factor for secondary lymphedema in cancer patients exposed to surgery or radiation. The risk is presumed to relate to its cytotoxicity. However, the present study provides initial evidence that doxorubicin directly inhibits lymph flow and this action appears distinct from its cytotoxic activity. We used real-time edge detection to track diameter changes in isolated rat mesenteric lymph vessels. Doxorubicin (0.5-20 µmol/l) progressively constricted lymph vessels and inhibited rhythmic contractions, reducing flow to 24.2% ± 7.7% of baseline. The inhibition of rhythmic contractions by doxorubicin paralleled a tonic rise in cytosolic Ca2+ concentration in lymphatic muscle cells, which was prevented by pharmacological antagonism of ryanodine receptors. Washout of doxorubicin partially restored lymph vessel contractions, implying a pharmacological effect. Subsequently, high-speed optical imaging was used to assess the effect of doxorubicin on rat mesenteric lymph flow in vivo. Superfusion of doxorubicin (0.05-10 µmol/l) maximally reduced volumetric lymph flow to 34% ± 11.6% of baseline. Likewise, doxorubicin (10 mg/kg) administered intravenously to establish clinically achievable plasma concentrations also maximally reduced volumetric lymph flow to 40.3% ± 6.0% of initial values. Our findings reveal that doxorubicin at plasma concentrations achieved during chemotherapy opens ryanodine receptors to induce "calcium leak" from the sarcoplasmic reticulum in lymphatic muscle cells and reduces lymph flow, an event linked to lymph vessel damage and the development of lymphedema. These results infer that pharmacological block of ryanodine receptors in lymphatic smooth muscle cells may mitigate secondary lymphedema in cancer patients subjected to doxorubicin chemotherapy. SIGNIFICANCE STATEMENT: Doxorubicin directly inhibits the rhythmic contractions of collecting lymph vessels and reduces lymph flow as a possible mechanism of secondary lymphedema, which is associated with the administration of anthracycline-based chemotherapy. The inhibitory effects of doxorubicin on rhythmic contractions and flow in isolated lymph vessels were prevented by pharmacological block of ryanodine receptors, thereby identifying the ryanodine receptor family of proteins as potential therapeutic targets for the development of new antilymphedema medications.


Assuntos
Doxorrubicina/farmacologia , Linfa/metabolismo , Vasos Linfáticos/metabolismo , Células Musculares/metabolismo , Contração Muscular/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Relação Dose-Resposta a Droga , Linfa/efeitos dos fármacos , Vasos Linfáticos/efeitos dos fármacos , Masculino , Células Musculares/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
7.
Microcirculation ; 25(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29072364

RESUMO

Voltage-gated K+ (Kv ) channels are major determinants of membrane potential in vascular smooth muscle cells (VSMCs) and regulate the diameter of small cerebral arteries and arterioles. However, the intracellular structures that govern the expression and function of vascular Kv channels are poorly understood. Scaffolding proteins including postsynaptic density 95 (PSD95) recently were identified in rat cerebral VSMCs. Primarily characterized in neurons, the PSD95 scaffold has more than 50 known binding partners, and it can mediate macromolecular signaling between cell-surface receptors and ion channels. In cerebral arteries, Shaker-type Kv 1 channels appear to associate with the PSD95 molecular scaffold, and PSD95 is required for the normal expression and vasodilator influence of members of this K+ channel gene family. Furthermore, recent findings suggest that the ß1-subtype adrenergic receptor is expressed in cerebral VSMCs and forms a functional vasodilator complex with Kv 1 channels on the PSD95 scaffold. Activation of ß1-subtype adrenergic receptors in VSMCs enables protein kinase A-dependent phosphorylation and opening of Kv 1 channels in the PSD95 complex; the subsequent K+ efflux mediates membrane hyperpolarization and vasodilation of small cerebral arteries. Early evidence from other studies suggests that other families of Kv channels and scaffolding proteins are expressed in VSMCs. Future investigations into these macromolecular complexes that modulate the expression and function of Kv channels may reveal unknown signaling cascades that regulate VSMC excitability and provide novel targets for ion channel-based medications to optimize vascular tone.


Assuntos
Circulação Cerebrovascular , Proteína 4 Homóloga a Disks-Large/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Receptores Adrenérgicos beta/fisiologia , Animais , Proteína 4 Homóloga a Disks-Large/metabolismo , Humanos , Músculo Liso Vascular/química , Músculo Liso Vascular/citologia , Ratos , Receptores Adrenérgicos beta/metabolismo
8.
Circ Res ; 114(8): 1258-67, 2014 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24585759

RESUMO

RATIONALE: Postsynaptic density-95 (PSD95) is a scaffolding protein that associates with voltage-gated, Shaker-type K(+) (KV1) channels and promotes the expression of KV1 channels in vascular smooth muscle cells of the cerebral (cVSMCs) circulation. However, the physiological role of PSD95 in mediating molecular signaling in cVSMCs is unknown. OBJECTIVE: We explored whether a specific interaction between PSD95 and KV1 channels enables protein kinase A phosphorylation of KV1 channels in cVSMCs to promote vasodilation. METHODS AND RESULTS: Rat cerebral arteries were used for analyses. A membrane-permeable peptide (KV1-C peptide) corresponding to the postsynaptic density-95, discs large, zonula occludens-1 binding motif in the C terminus of KV1.2α was designed as a dominant-negative peptide to disrupt the association of KV1 channels with PSD95. Application of KV1-C peptide to cannulated, pressurized cerebral arteries rapidly induced vasoconstriction and depolarized cVSMCs. These events corresponded to reduced coimmunoprecipitation of the PSD95 and KV1 proteins without altering surface expression. Middle cerebral arterioles imaged in situ through cranial window also constricted rapidly in response to local application of KV1-C peptide. Patch-clamp recordings confirmed that KV1-C peptide attenuates KV1 channel blocker (5-(4-phenylalkoxypsoralen))-sensitive current in cVSMCs. Western blots using a phospho-protein kinase A substrate antibody revealed that cerebral arteries exposed to KV1-C peptide showed markedly less phosphorylation of KV1.2α subunits. Finally, phosphatase inhibitors blunted both KV1-C peptide-mediated and protein kinase A inhibitor peptide-mediated vasoconstriction. CONCLUSIONS: These findings provide initial evidence that protein kinase A phosphorylation of KV1 channels is enabled by a dynamic association with PSD95 in cerebral arteries and suggest that a disruption of such association may compromise cerebral vasodilation and blood flow.


Assuntos
Artérias Cerebrais/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Potenciais da Membrana/fisiologia , Proteínas de Membrana/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large , Inibidores Enzimáticos/farmacologia , Masculino , Modelos Animais , Técnicas de Patch-Clamp , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Vasoconstrição/fisiologia , Vasodilatação/fisiologia
9.
Mol Pharmacol ; 83(2): 429-38, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23188715

RESUMO

Seizures are the manifestation of highly synchronized burst firing of a large population of cortical neurons. Epileptiform bursts with an underlying plateau potential in neurons are a cellular correlate of seizures. Emerging evidence suggests that the plateau potential is mediated by neuronal canonical transient receptor potential (TRPC) channels composed of members of the TRPC1/4/5 subgroup. We previously showed that TRPC1/4 double-knockout (DKO) mice lack epileptiform bursting in lateral septal neurons and exhibit reduced seizure-induced neuronal cell death, but surprisingly have unaltered pilocarpine-induced seizures. Here, we report that TRPC5 knockout (KO) mice exhibit both significantly reduced seizures and minimal seizure-induced neuronal cell death in the hippocampus. Interestingly, epileptiform bursting induced by agonists for metabotropic glutamate receptors in the hippocampal CA1 area is unaltered in TRPC5 KO mice, but is abolished in TRPC1 KO and TRPC1/4 DKO mice. In contrast, long-term potentiation is greatly reduced in TRPC5 KO mice, but is normal in TRPC1 KO and TRPC1/4 DKO mice. The distinct changes from these knockouts suggest that TRPC5 and TRPC1/4 contribute to seizure and excitotoxicity by distinct cellular mechanisms. Furthermore, the reduced seizure and excitotoxicity and normal spatial learning exhibited in TRPC5 KO mice suggest that TRPC5 is a promising novel molecular target for new therapy.


Assuntos
Região CA1 Hipocampal/patologia , Neurônios/fisiologia , Convulsões/metabolismo , Convulsões/patologia , Canais de Cátion TRPC/metabolismo , Animais , Região CA1 Hipocampal/metabolismo , Morte Celular/genética , Morte Celular/fisiologia , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Pilocarpina/farmacologia , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Convulsões/genética , Comportamento Espacial/fisiologia , Canais de Cátion TRPC/genética
10.
Pharmacol Res ; 70(1): 126-38, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23376354

RESUMO

Ion channels are multimeric, transmembrane proteins that selectively mediate ion flux across the plasma membrane in a variety of cells including vascular smooth muscle cells (VSMCs). The dynamic interplay of Ca(2+) and K(+) channels on the plasma membrane of VSMCs plays a pivotal role in modulating the vascular tone of small arteries and arterioles. The abnormally-elevated arterial tone observed in hypertension thus points to an aberrant expression and function of Ca(2+) and K(+) channels in the VSMCs. In this short review, we focus on the three well-studied ion channels in VSMCs, namely the L-type Ca(2+) (CaV1.2) channels, the voltage-gated K(+) (KV) channels, and the large-conductance Ca(2+)-activated K(+) (BK) channels. First, we provide a brief overview on the physiological role of vascular CaV1.2, KV and BK channels in regulating arterial tone. Second, we discuss the current understanding of the expression changes and regulation of CaV1.2, KV and BK channels in the vasculature during hypertension. Third, based on available proof-of-concept studies, we describe the potential therapeutic approaches targeting these vascular ion channels in order to restore blood pressure to normotensive levels.


Assuntos
Anti-Hipertensivos/farmacologia , Canais de Cálcio/metabolismo , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Canais de Potássio/metabolismo , Animais , Canais de Cálcio/genética , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Terapia de Alvo Molecular , Músculo Liso Vascular/metabolismo , Canais de Potássio/genética , Subunidades Proteicas
11.
Res Pract Thromb Haemost ; 7(2): 100058, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36865905

RESUMO

Background: Puncture wounding is a longstanding challenge to human health for which understanding is limited, in part, by a lack of detailed morphological data on how the circulating platelet capture to the vessel matrix leads to sustained, self-limiting platelet accumulation. Objectives: The objective of this study was to produce a paradigm for self-limiting thrombus growth in a mouse jugular vein model. Methods: Data mining of advanced electron microscopy images was performed from authors' laboratories. Results: Wide-area transmission electron mcrographs revealed initial platelet capture to the exposed adventitia resulted in localized patches of degranulated, procoagulant-like platelets. Platelet activation to a procoagulant state was sensitive to dabigatran, a direct-acting PAR receptor inhibitor, but not to cangrelor, a P2Y12 receptor inhibitor. Subsequent thrombus growth was sensitive to both cangrelor and dabigatran and sustained by the capture of discoid platelet strings first to collagen-anchored platelets and later to loosely adherent peripheral platelets. Spatial examination indicated that staged platelet activation resulted in a discoid platelet tethering zone that was pushed progressively outward as platelets converted from one activation state to another. As thrombus growth slowed, discoid platelet recruitment became rare and loosely adherent intravascular platelets failed to convert to tightly adherent platelets. Conclusions: In summary, the data support a model that we term Capture and Activate, in which the initial high platelet activation is directly linked to the exposed adventitia, all subsequent tethering of discoid platelets is to loosely adherent platelets that convert to tightly adherent platelets, and self-limiting, intravascular platelet activation over time is the result of decreased signaling intensity.

12.
Circ Res ; 106(4): 739-47, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20044515

RESUMO

RATIONALE: Calcium channel blockers (CCBs) exert their antihypertensive effect by reducing cardiac afterload but not preload, suggesting that Ca(2+) influx through L-type Ca(2+) channels (LTCC) mediates arterial but not venous tone. OBJECTIVE: The object of this study was to resolve the mechanism of venous resistance to CCBs. METHODS AND RESULTS: We compared the sensitivity of depolarization (KCl)-induced constriction of rat small mesenteric arteries (MAs) and veins (MVs) to the dilator effect of CCBs. Initial findings confirmed that nifedipine progressively dilated depolarization-induced constrictions in MAs but not MVs. However, Western blots showed a similar expression of the alpha(1C) pore-forming subunit of the LTCC in both vessels. Patch-clamp studies revealed a similar density of whole-cell Ca(2+) channel current between single smooth muscle cells (SMCs) of MAs and MVs. Based on these findings, we hypothesized that LTCCs are expressed but "silenced" by intracellular Ca(2+) in venous SMCs. After depletion of intracellular Ca(2+) stores by the SERCA pump inhibitor thapsigargin, depolarization-induced constrictions in MVs were blocked 80% by nifedipine suggesting restoration of Ca(2+) influx through LTCCs. Similarly, KCl-induced constrictions were sensitive to block by nifedipine after depletion of intracellular Ca(2+) stores by caffeine, ryanodine, or 2-aminoethoxydiphenyl borate. Cell-attached patch recordings of unitary LTCC currents confirmed rare channel openings during depolarization of venous compared to arterial SMCs, but chelating intracellular Ca(2+) significantly increased the open-state probability of venous LTCCs. CONCLUSIONS: We report that intracellular Ca(2+) inactivates LTCCs in venous SMCs to confer venous resistance to CCB-induced dilation, a fundamental drug property that was previously unexplained.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Cálcio/metabolismo , Resistência a Medicamentos , Músculo Liso Vascular/efeitos dos fármacos , Nifedipino/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Western Blotting , Canais de Cálcio Tipo L/metabolismo , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Masculino , Potenciais da Membrana , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Veias Mesentéricas/efeitos dos fármacos , Veias Mesentéricas/metabolismo , Músculo Liso Vascular/metabolismo , Técnicas de Patch-Clamp , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Vasoconstrição , Vasoconstritores/farmacologia , Vasodilatadores/metabolismo
13.
J Physiol ; 589(Pt 21): 5143-52, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21911612

RESUMO

Postsynaptic density-95 (PSD95) is a 95 kDa scaffolding molecule in the brain that clusters postsynaptic proteins including ion channels, receptors, enzymes and other signalling partners required for normal cognition. The voltage-gated, Shaker-type K(+) (K(V)1) channel is one key binding partner of PSD95 scaffolds in neurons. However, K(V)1 channels composed of α1.2 and α1.5 pore-forming subunits also are expressed in the vascular smooth muscle cells (cVSMCs) of the cerebral circulation, although the identity of their molecular scaffolds is unknown. Since α1.2 contains a binding motif for PSD95, we explored the possibility that cVSMCs express PSD95 as a scaffold to promote K(V)1 channel expression and cerebral vasodilatation. Cerebral arteries from Sprague-Dawley rats were isolated for analysis of PSD95 and K(V)1 channel proteins. PSD95 was detected in cVSMCs and it co-immunoprecipitated and co-localized with the pore-forming α1.2 subunit of the K(V)1 channel. Antisense-mediated knockdown of PSD95 profoundly reduced K(V)1 channel expression and suppressed K(V)1 current in patch-clamped cVSMCs. Loss of PSD95 also depolarized cVSMCs in pressurized cerebral arteries and induced a strong constriction associated with a loss of functional K(V)1 channels. Our findings provide initial evidence that PSD95 is expressed in cVSMCs, and the K(V)1 channel is one of its important binding partners. PSD95 appears to function as a critical 'dilator' scaffold in cerebral arteries by increasing the number of functional K(V)1 channels at the plasma membrane.


Assuntos
Artérias Cerebrais/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Miócitos de Músculo Liso/fisiologia , Densidade Pós-Sináptica/fisiologia , Superfamília Shaker de Canais de Potássio/fisiologia , Vasodilatação , Animais , Western Blotting , Proteína 4 Homóloga a Disks-Large , Furocumarinas/farmacologia , Técnicas de Silenciamento de Genes , Masculino , Potenciais da Membrana/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Venenos de Aranha/farmacologia , Vasodilatação/efeitos dos fármacos
14.
Am J Physiol Renal Physiol ; 301(1): F209-17, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21511700

RESUMO

Sepsis is a leading cause of acute kidney injury (AKI) and mortality in children. Understanding the development of pediatric sepsis and its effects on the kidney are critical in uncovering new therapies. The goal of this study was to characterize the development of sepsis-induced AKI in the clinically relevant cecal ligation and puncture (CLP) model of peritonitis in rat pups 17-18 days old. CLP produced severe sepsis demonstrated by time-dependent increase in serum cytokines, NO, markers of multiorgan injury, and renal microcirculatory hypoperfusion. Although blood pressure and heart rate remained unchanged after CLP, renal blood flow (RBF) was decreased 61% by 6 h. Renal microcirculatory analysis showed the number of continuously flowing cortical capillaries decreased significantly from 69 to 48% by 6 h with a 66% decrease in red blood cell velocity and a 57% decline in volumetric flow. The progression of renal microcirculatory hypoperfusion was associated with peritubular capillary leakage and reactive nitrogen species generation. Sham adults had higher mean arterial pressure (118 vs. 69 mmHg), RBF (4.2 vs. 1.1 ml·min(-1)·g(-1)), and peritubular capillary velocity (78% continuous flowing capillaries vs. 69%) compared with pups. CLP produced a greater decrease in renal microcirculation in pups, supporting the notion that adult models may not be the most appropriate for studying pediatric sepsis-induced AKI. Lower RBF and reduced peritubular capillary perfusion in the pup suggest the pediatric kidney may be more susceptible to AKI than would be predicted using adults models.


Assuntos
Injúria Renal Aguda/fisiopatologia , Hemodinâmica/fisiologia , Circulação Renal/fisiologia , Sepse/fisiopatologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Animais Recém-Nascidos , Biomarcadores/sangue , Velocidade do Fluxo Sanguíneo/fisiologia , Pressão Sanguínea/fisiologia , Permeabilidade Capilar/fisiologia , Ceco/fisiologia , Hidratação , Hipotermia/etiologia , Hipotermia/fisiopatologia , Imuno-Histoquímica , Rim/patologia , Ligadura , Masculino , Microcirculação/fisiologia , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Peritonite/etiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/complicações , Telemetria
15.
Commun Biol ; 4(1): 1090, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34531522

RESUMO

Primary hemostasis results in a platelet-rich thrombus that has long been assumed to form a solid plug. Unexpectedly, our 3-dimensional (3D) electron microscopy of mouse jugular vein puncture wounds revealed that the resulting thrombi were structured about localized, nucleated platelet aggregates, pedestals and columns, that produced a vaulted thrombus capped by extravascular platelet adherence. Pedestal and column surfaces were lined by procoagulant platelets. Furthermore, early steps in thrombus assembly were sensitive to P2Y12 inhibition and late steps to thrombin inhibition. Based on these results, we propose a Cap and Build, puncture wound paradigm that should have translational implications for bleeding control and hemostasis.


Assuntos
Plaquetas/fisiologia , Hemostasia/fisiologia , Punções/efeitos adversos , Trombose/fisiopatologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Trombose/etiologia
16.
Am J Physiol Heart Circ Physiol ; 297(1): H293-303, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19411284

RESUMO

The cerebral arteries of hypertensive rats are depolarized and highly myogenic, suggesting a loss of K(+) channels in the vascular smooth muscle cells (VSMCs). The present study evaluated whether the dilator function of the prominent Shaker-type voltage-gated K(+) (K(V)1) channels is attenuated in middle cerebral arteries from two rat models of hypertension. Block of K(V)1 channels by correolide (1 micromol/l) or psora-4 (100 nmol/l) reduced the resting diameter of pressurized (80 mmHg) cerebral arteries from normotensive rats by an average of 28 +/- 3% or 26 +/- 3%, respectively. In contrast, arteries from spontaneously hypertensive rats (SHR) and aortic-banded (Ao-B) rats with chronic hypertension showed enhanced Ca(2+)-dependent tone and failed to significantly constrict to correolide or psora-4, implying a loss of K(V)1 channel-mediated vasodilation. Patch-clamp studies in the VSMCs of SHR confirmed that the peak K(+) current density attributed to K(V)1 channels averaged only 5.47 +/- 1.03 pA/pF, compared with 9.58 +/- 0.82 pA/pF in VSMCs of control Wistar-Kyoto rats. Subsequently, Western blots revealed a 49 +/- 7% to 66 +/- 7% loss of the pore-forming alpha(1.2)- and alpha(1.5)-subunits that compose K(V)1 channels in cerebral arteries of SHR and Ao-B rats compared with control animals. In each case, the deficiency of K(V)1 channels was associated with reduced mRNA levels encoding either or both alpha-subunits. Collectively, these findings demonstrate that a deficit of alpha(1.2)- and alpha(1.5)-subunits results in a reduced contribution of K(V)1 channels to the resting diameters of cerebral arteries from two rat models of hypertension that originate from different etiologies.


Assuntos
Hipertensão Renovascular/genética , Hipertensão Renovascular/fisiopatologia , Hipertensão/genética , Hipertensão/fisiopatologia , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/fisiologia , Vasodilatação/genética , Vasodilatação/fisiologia , Acetilcolina/farmacologia , Animais , Aorta/fisiologia , Western Blotting , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiologia , Masculino , Técnicas de Patch-Clamp , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serotonina/farmacologia , Vasodilatadores/farmacologia
17.
J Pharmacol Exp Ther ; 329(2): 775-82, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19244098

RESUMO

In different rodent models of hypertension, vascular voltage-gated L-type calcium channel (Ca(L)) current and vascular tone is increased because of increased expression of the noncardiac form of the Ca(L) (Ca(v)1.2). The objective of this study was to develop a small interfering RNA (siRNA) expression system against the noncardiac form of Ca(v)1.2 to reduce its expression in vascular smooth muscle cells (VSMCs). siRNAs expressing plasmids and appropriate controls were constructed and first screened in human embryonic kidney (HEK) 293 cells cotransfected with a rat Ca(v)1.2 expression vector. The most effective gene silencing was achieved with a modified mir-30a-based short hairpin RNA (shRNAmir) driven by the cytomegalovirus promoter. In A7r5 cells, a vascular smooth muscle cell line, two copies of shRNAmir driven by a chimeric VSMC-specific enhancer/promoter reduced endogenous Ca(v)1.2 expression by 61% and decreased the Ca(L) current carried by barium by 47%. Moreover, the chimeric vascular smooth muscle-specific enhancer/promoter displayed almost no activity in non-VSMCs (PC-12 and HEK 293). Because the proposed siRNA was designed to only target the noncardiac form of Ca(v)1.2, it did not affect the Ca(L) expression and function in cultured cardiomyocytes, even when driven by a stronger cytomegalovirus promoter. In conclusion, vascular Ca(v)1.2 expression and function were effectively reduced by VSMC-specific delivery of the noncardiac form of Ca(v)1.2 siRNA without similarly affecting cardiac Ca(L) expression and function. When coupled with a viral vector, this molecular intervention in vivo may provide a novel long-term vascular-specific gene therapy for hypertension.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Hipertensão/terapia , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Animais , Western Blotting , Canais de Cálcio Tipo L/genética , Linhagem Celular , Regulação para Baixo , Terapia Genética , Humanos , Hipertensão/genética , Hipertensão/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Técnicas de Patch-Clamp , Plasmídeos , Ratos , Transfecção
18.
Mol Biol Cell ; 17(9): 4105-17, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16837554

RESUMO

In yeast, particular emphasis has been given to endoplasmic reticulum (ER)-derived, cisternal maturation models of Golgi assembly while in mammalian cells more emphasis has been given to golgins as a potentially stable assembly framework. In the case of de novo Golgi formation from the ER after brefeldin A/H89 washout in HeLa cells, we found that scattered, golgin-enriched, structures formed early and contained golgins including giantin, ranging across the entire cis to trans spectrum of the Golgi apparatus. These structures were incompetent in VSV-G cargo transport. Second, we compared Golgi competence in cargo transport to the kinetics of addition of various glycosyltransferases and glycosidases into nascent, golgin-enriched structures after drug washout. Enzyme accumulation was sequential with trans and then medial glycosyltransferases/glycosidases found in the scattered, nascent Golgi. Involvement in cargo transport preceded full accumulation of enzymes or GPP130 into nascent Golgi. Third, during mitosis, we found that the formation of a golgin-positive acceptor compartment in early telophase preceded the accumulation of a Golgi glycosyltransferase in nascent Golgi structures. We conclude that during mammalian Golgi assembly components fit into a dynamic, first-formed, multigolgin-enriched framework that is initially cargo transport incompetent. Resumption of cargo transport precedes full Golgi assembly.


Assuntos
Complexo de Golgi/metabolismo , Transporte Biológico/efeitos dos fármacos , Brefeldina A/farmacologia , Células Cultivadas , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Glicosídeo Hidrolases/metabolismo , Glicosiltransferases/metabolismo , Complexo de Golgi/efeitos dos fármacos , Células HeLa , Humanos , Isoquinolinas/farmacologia , Cinética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Mitose/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Sulfonamidas/farmacologia , Proteínas do Envelope Viral/metabolismo
19.
J Pharmacol Exp Ther ; 325(1): 37-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18184831

RESUMO

Calcium influx through long-lasting ("L-type") Ca(2+) channels (Ca(V)) drives excitation-contraction in the normal heart. Dysregulation of this process contributes to Ca(2+) overload, and interventions that reduce expression of the pore-forming alpha(1) subunit may alleviate cytosolic Ca(2+) excess. As a molecular approach to disrupt the assembly of Ca(V)1.2 (alpha(1C)) channels at the cell membrane, we targeted the Ca(2+) channel beta(2) subunit, an intracellular chaperone that interacts with alpha(1C) via its beta interaction domain (BID) to promote Ca(V)1.2 channel expression. Recombinant adenovirus expressing either the full beta(2) subunit (Full-beta(2)) or truncated beta(2) subunit constructs lacking either the C terminus, N terminus, or both (N-BID, C-BID, and BID, respectively) fused to green fluorescent protein were developed as potential decoys and overexpressed in HL-1 cells. Fluorescence microscopy revealed that the localization of Full-beta(2) at the surface membrane was associated with increased Ca(2+) current mainly attributed to Ca(V)1.2 channels. In contrast, truncated N-BID and C-BID constructs showed punctate intracellular expression, and BID showed a diffuse cytosolic distribution. Total expression of the alpha(1C) protein of Ca(V)1.2 channels was similar between groups, but HL-1 cells overexpressing C-BID and BID exhibited reduced Ca(2+) current. C-BID and BID also attenuated Ca(2+) current associated with another L-type Ca(2+) channel, Ca(V)1.3, but they did not reduce transient Ca(2+) currents attributed to Ca(V)3 channels. These results suggest that beta(2) subunit mutants lacking the N terminus may preferentially disrupt the proper localization of L-type Ca(2+) channels in the cell membrane. Cardiac-specific delivery of these decoy molecules in vivo may represent a gene-based treatment for pathologies involving Ca(2+) overload.


Assuntos
Canais de Cálcio Tipo L/efeitos dos fármacos , Desenho de Fármacos , Terapia Genética/métodos , Mimetismo Molecular , Mutação , Miocárdio/química , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular , DNA Complementar/genética , Regulação para Baixo/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Humanos , Miocárdio/citologia , Engenharia de Proteínas/métodos , Subunidades Proteicas/genética , Subunidades Proteicas/uso terapêutico , Ratos
20.
Trans Am Clin Climatol Assoc ; 119: 171-82; discussion 182-3, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18596857

RESUMO

Modifying ion channel expression and function in the heart and vasculature are potentially useful, novel approaches to managing cardiac hypertrophy, atrial fibrillation and hypertension. Calcium channels play a pivotal role in the heart and vasculature in controlling muscle contraction as well as other aspects of calcium-dependent signaling. The present investigation reports development of mutated L-type calcium channel beta subunits that are delivered by an adenoviral vector to vascular smooth muscle tissue. Wild type subunits serve a chaperone function for the pore-forming alpha(1C) subunit of the calcium channel, localize to the cell membrane and enhance calcium current. Conversely, mutated subunits function as dominant negative, defective chaperone molecules that disrupt targeting to the cell membrane and decrease calcium current. The dominant negative genes can be delivered in vitro and ex vivo, and have the potential to decrease arterial tone and lower blood pressure in vivo.


Assuntos
Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Terapia Genética/métodos , Canais Iônicos/genética , Animais , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/fisiologia , Doenças Cardiovasculares/fisiopatologia , Células Cultivadas , Vetores Genéticos , Hipertensão/genética , Hipertensão/fisiopatologia , Hipertensão/terapia , Canais Iônicos/fisiologia , Artérias Mesentéricas/citologia , Artérias Mesentéricas/fisiologia , Miócitos de Músculo Liso/fisiologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA