Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Regul Integr Comp Physiol ; 323(5): R763-R775, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36189990

RESUMO

Migration of monocytes-macrophages plays an important role in phagocytosis of pathogens and cellular debris in a variety of pathophysiological conditions. Although epithelial Na+ channels (ENaCs) are required for normal migratory responses in other cell types, their role in macrophage migration signaling is unknown. To address this possibility, we determined whether ENaC message is present in several peripheral blood monocyte cell populations and tissue-resident macrophages in healthy humans using the Human Protein Atlas database (www.proteinatlas.org) and the mouse monocyte cell line RAW 264.7 using RT-PCR. We then determined that selective ENaC inhibition with amiloride inhibited chemotactic migration (∼50%), but not phagocytosis, of the mouse monocyte-macrophage cell line RAW 264.7. Furthermore, we generated a cell line stably expressing an NH2-terminal truncated αENaC to interrupt normal channel trafficking and found it suppressed migration. Prolonged exposure (48 h) of RAW 264.7 cells to proinflammatory cytokines interferon γ (IFNγ) and/or tumor necrosis factor α (TNFα) inhibited RAW 264.7 migration and abolished the amiloride (1 µM)-sensitive component of migration, a finding consistent with ENaC downregulation. To determine if proinflammatory cytokines regulate αENaC protein expression, cells were exposed to proinflammatory cytokines IFNγ (10 ng/mL, last 48 h) and TNFα (10 ng/mL, last 24 h). By Western blot analysis, we found whole cell αENaC protein is reduced ≥50%. Immunofluorescence demonstrated heterogeneous αENaC inhibition. Finally, we found that overnight exposure to amiloride stimulated morphological changes and increased polarization marker expression. Our findings suggest that ENaC may be a critical molecule in macrophage migration and polarization.


Assuntos
Canais Epiteliais de Sódio , Fator de Necrose Tumoral alfa , Camundongos , Animais , Humanos , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Amilorida/farmacologia , Interferon gama/farmacologia , Interferon gama/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo
2.
Physiol Rep ; 10(13): e15376, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35831968

RESUMO

Preeclampsia (PE) is associated with adverse cerebrovascular effects during and following parturition including stroke, small vessel disease, and vascular dementia. A potential contributing factor to the cerebrovascular dysfunction is the loss of cerebral blood flow (CBF) autoregulation. Autoregulation is the maintenance of CBF to meet local demands with changes in perfusion pressure. When perfusion pressure rises, vasoconstriction of cerebral arteries and arterioles maintains flow and prevents the transfer of higher systemic pressure to downstream microvasculature. In the face of concurrent hypertension, loss of autoregulatory control exposes small delicate microvessels to injury from elevated systemic blood pressure. While placental ischemia is considered the initiating event in the preeclamptic cascade, the factor(s) mediating cerebrovascular dysfunction are poorly understood. Elevated plasma proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin-17 (IL-17), are potential mediators of autoregulatory loss. Impaired CBF responses to increases in systemic pressure are attributed to the impaired pressure-induced (myogenic) constriction of small cerebral arteries and arterioles in PE. Myogenic vasoconstriction is initiated by pressure-induced vascular smooth muscle cell (VSMC) stretch. Recent studies from our laboratory group indicate that proinflammatory cytokines impair the myogenic mechanism of CBF autoregulation via inhibition of vascular degenerin proteins, putative mediators of myogenic constriction in VSMCs. This brief review links studies showing the effect of proinflammatory cytokines on degenerin expression and CBF autoregulation to the pathological cerebral consequences of preeclampsia.


Assuntos
Pré-Eclâmpsia , Circulação Cerebrovascular/fisiologia , Citocinas/farmacologia , Canais de Sódio Degenerina , Feminino , Humanos , Placenta , Gravidez
3.
Am J Physiol Renal Physiol ; 322(5): F498-F511, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35285274

RESUMO

In independent studies, our laboratory has shown the importance of the degenerin proteins ß-epithelial Na+ channel (ßENaC) and acid-sensing ion channel 2 (ASIC2) in pressure-induced constriction (PIC) in renal interlobar arteries. Most, but not all, of the PIC response is abolished in mice lacking normal levels of ßENaC or in ASIC2-null mice, indicating that the functions of ßENaC and ASIC2 cannot fully compensate for the loss of the other. Degenerin proteins are known to associate and form heteromeric channels in expression systems, but whether they interact biochemically and functionally in vascular smooth muscle cells is unknown. We hypothesized that ßENaC and ASIC2 interact to mediate PIC responses in renal vessels. To address this possibility, we 1) used biochemical approaches to show that ßENaC associates into high-molecular-weight complexes and immunoprecipitants with ASIC2 in vascular smooth muscle cells and then 2) examined PIC in renal afferent arterioles in mice lacking normal levels of ßENaC (ßENaCm/m) or/and ASIC2 (ASIC2-/-) using the isolated afferent arteriole-attached glomerulus preparation. We found that the sensitivity of the PIC response (slope of the relationship between intraluminal pressure and percent myogenic tone) decreased to 26%, 27%, and -8% of wild-type controls in ASIC2-/-, ßENaCm/m, and ASIC2-/-/ßENaCm/m groups, respectively, suggesting that the PIC response was totally abolished in mice deficient in both ASIC2 and ßENaC. Surprisingly, we found that resting internal diameters were 20-30% lower (60 mmHg, Ca2+ free) in ASIC2-/-/ßENaCm/m (11.3 ± 0.5 µm) mice compared with control (14.4 ± 0.6 µm, P = 0.0007, independent two-tailed t test) or singly modified (15.7 ± 1.0 to 16.3 ± 1.1 µm) mice, suggesting compensatory vasoconstriction or remodeling. We then examined mean arterial blood pressure (MAP) using radiotelemetry and glomerular injury using histological examination of renal sections. We found that 24-h MAP was mildly elevated (+8 mmHg) in ASIC2-/-/ßENaCm/m mice versus wild-type controls and the glomerular injury score was modestly increased by 38%. These findings demonstrate that myogenic constriction in afferent arterioles is dependent on normal expression of ßENaC and ASIC2 and that mice lacking normal levels of ASIC2 and ßENaC have mild renal injury and increased MAP.NEW & NOTEWORTHY Transmission of systemic blood pressure to delicate renal microvessels is a primary determinant of vascular injury in chronic kidney disease progression to end-stage renal disease. Here, we identified two degenerin family members, with an evolutionary link to mechanosensing, that interact biochemically and functionally to regulate systemic blood pressure and renal injury. Thus, degenerin proteins may serve as a target for the development of therapies to prevent or delay renal disease progression.


Assuntos
Rim , Músculo Liso Vascular , Animais , Arteríolas , Constrição , Camundongos , Músculo Liso Vascular/metabolismo , Vasoconstrição
4.
Physiology (Bethesda) ; 36(5): 292-306, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34431420

RESUMO

Loss of pressure-induced vasoconstriction increases susceptibility to renal and cerebral vascular injury. Favored paradigms underlying initiation of the response include transient receptor potential channels coupled to G protein-coupled receptors or integrins as transducers. Degenerin channels may also mediate the response. This review addresses the 1) evolutionary role of these molecules in mechanosensing, 2) limitations to identifying mechanosensitive molecules, and 3) paradigm shifting molecular model for a VSMC mechanosensor.


Assuntos
Músculo Liso Vascular , Miócitos de Músculo Liso , Rim , Vasoconstrição
5.
Am J Hypertens ; 34(11): 1227-1235, 2021 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-34161569

RESUMO

BACKGROUND: Pressure-induced constriction (PIC) is inherent to small arteries and arterioles, in which intraluminal pressure-induced vascular smooth muscle cell stretch elicits vasoconstriction. Degenerin (Deg) proteins, such as beta-epithelial Na+ channel (ßENaC), have been studied in the PIC response because they are evolutionarily linked to known mechanosensors. While loss of Deg function phenotypes are plentiful, a gain-of-function phenotype has not been studied. The aim of this study was to determine if expression of exogenous ßENaC in the isolated middle cerebral artery (MCA) enhances the PIC response. METHODS: Isolated MCA segments from female mice (24 weeks, n = 5) were transfected with enhanced green fluorescent protein-ßENaC (EGFP-ßENaC) or with EGFP alone, incubated overnight at 37 °C, then studied in a pressure myograph. RESULTS: Mechanical/morphological properties and vasoconstrictor responses to KCl and phenylephrine were identical in EGFP-ßENaC and EGFP MCAs. In contrast, PIC responses were greater in EGFP-ßENaC segments with ~2-fold greater peak myogenic tone. CONCLUSIONS: These data confirm previous findings that ßENaC is critical in the PIC response. These data provide proof-of-concept that upregulating ßENaC can enhance PIC responses and lay the foundation to test the hypothesis that inflammation-mediated downregulation of ßENaC contributes to cerebrovascular dysfunction.


Assuntos
Canais Epiteliais de Sódio , Artéria Cerebral Média , Animais , Constrição , Canais de Sódio Degenerina/metabolismo , Canais Epiteliais de Sódio/genética , Feminino , Camundongos , Músculo Liso Vascular/metabolismo , Fenilefrina/farmacologia , Sódio/metabolismo , Vasoconstrição , Vasoconstritores/farmacologia
6.
Pregnancy Hypertens ; 24: 50-57, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33677419

RESUMO

Preeclampsia affects 5-8% of pregnancies and is characterized by hypertension, placental ischemia, neurological impairment, and an increase in circulating inflammatory cytokines, including Interleukin-17 (IL17). While placental ischemia has also been shown to impair cerebrovascular function, it is not known which placental-associated factor(s) drive this effect. The purpose of this study was to examine the effects of IL17 on cerebrovascular function during pregnancy. To achieve this goal, pregnant rats were infused with either IL17 (150 pg/day, 5 days, osmotic minipump), or vehicle (saline/0.7% BSA osmotic minipump) starting at gestational day (GD) 14. On GD 19, the cerebral blood flow (CBF) response to increases in mean arterial pressure (MAP) was measured in vivo, and myogenic constrictor responses of the middle cerebral artery (MCA) were assessed ex vivo. IL17 increased MAP but impaired CBF responses only at the highest arterial pressure measured (190 mmHg). Myogenic constrictor responses overall were mostly unaffected by IL17 infusion; however, the intraluminal pressure at which peak myogenic tone was generated was lower in the IL17 infused group (120 vs 165 mm Hg), suggesting maximal tone is exerted at lower intraluminal pressures in IL17-treated pregnant rats. Consistent with the lack of substantial change in overall myogenic responsiveness, there was no difference in cerebral vessel expression of putative mechanosensitive protein ßENaC, but a tendency towards a decrease in ASIC2 (p = 0.067) in IL17 rats. This study suggests that infusion of IL17 independent of other placental ischemia-associated factors is insufficient to recapitulate the features of impaired cerebrovascular function during placental ischemia. Further studies to examine of the role of other pro-inflammatory cytokines, individually or a combination, are necessary to determine mechanisms of cerebral vascular dysfunction during preeclampsia.


Assuntos
Circulação Cerebrovascular , Hipertensão/fisiopatologia , Interleucina-17/farmacologia , Artéria Cerebral Média/efeitos dos fármacos , Pré-Eclâmpsia/etiologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Canais Iônicos Sensíveis a Ácido/farmacologia , Animais , Pressão Sanguínea , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Interleucina-17/metabolismo , Artéria Cerebral Média/metabolismo , Gravidez , Ratos Sprague-Dawley
7.
Compr Physiol ; 11(1): 1315-1349, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33295016

RESUMO

Preeclampsia (PE), a hypertensive disorder, occurs in 3% to 8% of pregnancies in the United States and affects over 200,000 women and newborns per year. The United States has seen a 25% increase in the incidence of PE, largely owing to increases in risk factors, including obesity and cardiovascular disease. Although the etiology of PE is not clear, it is believed that impaired spiral artery remodeling of the placenta reduces perfusion, leading to placental ischemia. Subsequently, the ischemic placenta releases antiangiogenic and pro-inflammatory factors, such as cytokines, reactive oxygen species, and the angiotensin II type 1 receptor autoantibody (AT1-AA), among others, into the maternal circulation. These factors cause widespread endothelial activation, upregulation of the endothelin system, and vasoconstriction. In turn, these changes affect the function of multiple organ systems including the kidneys, brain, liver, and heart. Despite extensive research into the pathophysiology of PE, the only treatment option remains early delivery of the baby and importantly, the placenta. While premature delivery is effective in ameliorating immediate risk to the mother, mounting evidence suggests that PE increases risk of cardiovascular disease later in life for both mother and baby. Notably, these women are at increased risk of hypertension, heart disease, and stroke, while offspring are at risk of obesity, hypertension, and neurological disease, among other complications, later in life. This article aims to discuss the current understanding of the diagnosis and pathophysiology of PE, as well as associated organ damage, maternal and fetal outcomes, and potential therapeutic avenues. © 2021 American Physiological Society. Compr Physiol 11:1315-1349, 2021.


Assuntos
Hipertensão , Pré-Eclâmpsia , Feminino , Humanos , Recém-Nascido , Isquemia , Placenta , Pré-Eclâmpsia/epidemiologia , Gravidez , Fatores de Risco
8.
Am J Physiol Heart Circ Physiol ; 319(1): H42-H50, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32412783

RESUMO

Pressure-induced constriction (PIC) is an inherent response of small arteries and arterioles in which increases in intraluminal pressure evoke vasoconstriction. It is a critical mechanism of blood flow autoregulation in the kidney and brain. Degenerin (Deg) and transient receptor potential (Trp) protein families have been implicated in transduction of PIC because of evolutionary links to mechanosensing in the nematode and fly. While TrpC6 has been suggested to contribute to PIC signaling, direct supporting evidence is contradictory. Therefore, the aim of this study was to determine the importance of TrpC6 in PIC signaling using a mouse model lacking TrpC6. To address this aim, we evaluated graded pressure (20-90 mmHg), depolarization (4-80 mM KCl)-, and adrenergic receptor (phenylephrine; PE 10-7-10-4 M)-mediated constriction of isolated middle cerebral artery (MCA) segments from 9-wk-old male wild-type (TrpC6+/+, n = 7) and homozygous null (TrpC6-/-, n = 9) TrpC6 mice (Jackson Laboratories). Isolated MCA segments were cannulated and pressurized with physiological salt solution using pressure myography (Living Systems). Vasoconstrictor responses to KCl and PE were identical in TrpC6-/- and TrpC6+/+ mice. In contrast, PIC responses were totally abolished in TrpC6-/- mice. At 90 mmHg, the calculated myogenic tone was -0.8 ± 0.5 vs. 10.7 ± 1.7%, P = 0.0002 in TrpC6-/- and TrpC6+/+ mice, respectively. Additionally, there were no changes in mechanical properties of circumferential wall strain and stress or morphological properties of wall thickness and wall-to-lumen ratio at 50 mmHg between TrpPC6-/- and TrpC6+/+ mice. Although these results demonstrate that TrpC6 is critical for the integrated PIC response, they do not identify whether TrpC6 acts as a mechanosensor or a downstream signaling component.NEW & NOTEWORTHY Pressure-induced, but not agonist-induced, vasoconstriction is abolished in the middle cerebral artery (MCA) of TrpC6 null mice. TrpC6 localization in dissociated cerebral vascular smooth muscle cells is primarily cytoplasmic and not associated with the surface membrane where a mechanoelectrical coupler might be expected. These findings suggest that TrpC6 is required for transduction of pressure-induced constriction in the MCA; however, its role as a mechanoelectrical coupler or downstream signal amplifier remains unresolved.


Assuntos
Artéria Cerebral Média/metabolismo , Pressão , Canal de Cátion TRPC6/metabolismo , Vasoconstrição , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Artéria Cerebral Média/efeitos dos fármacos , Artéria Cerebral Média/fisiologia , Tono Muscular , Fenilefrina/farmacologia , Potássio/farmacologia , Canal de Cátion TRPC6/genética , Vasoconstritores/farmacologia
9.
Int J Mol Sci ; 21(8)2020 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-32331392

RESUMO

Degenerin proteins, such as the beta epithelial Na+ channel (ßENaC), are essential in the intracellular signaling of pressure-induced constriction, an important vascular smooth muscle cell (VSMC) function. While certain cytokines reduce ENaC protein in epithelial tissue, it is unknown if interleukin-17 (IL-17), a potent pro-inflammatory cytokine, directly mediates changes in membrane-associated ßENaC in VSMCs. Therefore, we tested the hypothesis that exposure to IL-17 reduces ßENaC in VSMCs through canonical mitogen-activated protein kinase (MAPK) signaling pathways. We treated cultured rat VSMCs (A10 cell line) with IL-17 (1-100 ng/mL) for 15 min to 16 h and measured expression of ßENaC, p38MAPK, c-jun kinase (JNK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). IL-17 reduced ßENaC protein expression in a concentration-dependent fashion and increased phosphorylation of p38MAPK by 15 min and JNK by 8 h. NFκB was unaffected by IL-17 in VSMCs. IL-17 treatment reduced VSMC viability but had no effect on cell death. To determine the underlying signaling pathway involved in this response, VSMCs were treated before and during IL-17 exposure with p38MAPK or JNK inhibitors. We found that JNK blockade prevented IL-17-mediated ßENaC protein suppression. These data demonstrate that the pro-inflammatory cytokine IL-17 regulates VSMC ßENaC via canonical MAPK signaling pathways, raising the possibility that ßENaC-mediated loss of VSMC function may occur in inflammatory disorders.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Interleucina-17/metabolismo , Sistema de Sinalização das MAP Quinases , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Canais Epiteliais de Sódio/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-17/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Fosforilação , Ratos
10.
Am J Physiol Heart Circ Physiol ; 318(4): H1018-H1027, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32167780

RESUMO

Preeclampsia is a pregnancy-related disorder characterized by hypertension, vascular dysfunction and an increase in circulating inflammatory factors including the cytokine, tumor necrosis factor-α (TNF-α). Studies have shown that placental ischemia is associated with 1) increased circulating TNF-α, 2) attenuated pressure-induced cerebral vascular tone, and 3) suppression of ß-epithelial Na+ channel (ßENaC) protein in cerebral vessels. In addition to its role in epithelial Na+ and water transport, ßENaC is an essential signaling element in transduction of pressure-induced (aka "myogenic") constriction, a critical mechanism of blood flow autoregulation. While cytokines inhibit expression of certain ENaC proteins in epithelial tissue, it is unknown if the increased circulating TNF-α associated with placental ischemia mediates the loss of cerebrovascular ßENaC and cerebral blood flow regulation. Therefore, the purpose of this study was to test the hypothesis that increasing plasma TNF-α in normal pregnant rats reduces cerebrovascular ßENaC expression and impairs cerebral blood flow (CBF) regulation. In vivo TNF-α infusion (200 ng/day, 5 days) inhibited cerebrovascular expression of ßENaC and impaired CBF regulation in pregnant rats. To determine the direct effects of TNF-α and underlying pathways mediating vascular smooth muscle cell ßENaC reduction, we exposed cultured VSMCs (A10 cell line) to TNF-α (1-100 ng/mL) for 16-24 h. TNF-α reduced ßENaC protein expression in a concentration-dependent fashion from 0.1 to 100 ng/mL, without affecting cell death. To assess the role of canonical MAPK signaling in this response, VSMCs were treated with p38MAPK or c-Jun kinase (JNK) inhibitors in the presence of TNF-α. We found that both p38MAPK and JNK blockade prevented TNF-α-mediated ßENaC protein suppression. These data provide evidence that disorders associated with increased circulating TNF-α could lead to impaired cerebrovascular regulation, possibly due to reduced ßENaC-mediated vascular function.NEW & NOTEWORTHY This manuscript identifies TNF-α as a possible placental-derived cytokine that could be involved in declining cerebrovascular health observed in preeclampsia. We found that infusion of TNF-α during pregnancy impaired cerebral blood flow control in rats at high arterial pressures. We further discovered that cerebrovascular ß-epithelial sodium channel (ßENaC) protein, a degenerin protein involved in mechanotransduction, was reduced by TNF-α in pregnant rats, indicating a potential link between impaired blood flow and this myogenic player. We next examined this effect in vitro using a rat vascular smooth muscle cell line. TNF-α reduced ßENaC through canonical MAPK-signaling pathways and was not dependent on cell death. This study demonstrates the pejorative effects of TNF-α on cerebrovascular function during pregnancy and warrants future investigations to study the role of cytokines on vascular function during pregnancy.


Assuntos
Circulação Cerebrovascular , Canais Epiteliais de Sódio/metabolismo , Músculo Liso Vascular/metabolismo , Pré-Eclâmpsia/etiologia , Fator de Necrose Tumoral alfa/sangue , Animais , Pressão Sanguínea , Linhagem Celular , Células Cultivadas , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Canais Epiteliais de Sódio/genética , Feminino , Homeostase , Sistema de Sinalização das MAP Quinases , Músculo Liso Vascular/efeitos dos fármacos , Gravidez , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/farmacologia
11.
Biol Sex Differ ; 10(1): 58, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31829239

RESUMO

BACKGROUND: Placental ischemia and hypertension, characteristic features of preeclampsia, are associated with impaired cerebral blood flow (CBF) autoregulation and cerebral edema. However, the factors that contribute to these cerebral abnormalities are not clear. Several lines of evidence suggest that angiotensin II can impact cerebrovascular function; however, the role of the renin angiotensin system in cerebrovascular function during placental ischemia has not been examined. We tested whether the angiotensin type 1 (AT1) receptor contributes to impaired CBF autoregulation in pregnant rats with placental ischemia caused by surgically reducing uterine perfusion pressure. METHODS: Placental ischemic or sham operated rats were treated with vehicle or losartan from gestational day (GD) 14 to 19 in the drinking water. On GD 19, we assessed CBF autoregulation in anesthetized rats using laser Doppler flowmetry. RESULTS: Placental ischemic rats had impaired CBF autoregulation that was attenuated by treatment with losartan. In addition, we examined whether an agonistic autoantibody to the AT1 receptor (AT1-AA), reported to be present in preeclamptic women, contributes to impaired CBF autoregulation. Purified rat AT1-AA or vehicle was infused into pregnant rats from GD 12 to 19 via mini-osmotic pumps after which CBF autoregulation was assessed. AT1-AA infusion impaired CBF autoregulation but did not affect brain water content. CONCLUSIONS: These results suggest that the impaired CBF autoregulation associated with placental ischemia is due, at least in part, to activation of the AT1 receptor and that the RAS may interact with other placental factors to promote cerebrovascular changes common to preeclampsia.


Assuntos
Circulação Cerebrovascular , Homeostase , Isquemia/fisiopatologia , Placenta/irrigação sanguínea , Receptor Tipo 1 de Angiotensina/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Feminino , Losartan/farmacologia , Pré-Eclâmpsia/fisiopatologia , Gravidez , Ratos Sprague-Dawley
12.
Antioxidants (Basel) ; 8(6)2019 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216709

RESUMO

Heme oxygenase (HO) plays an important role in the cardiovascular system. It is involved in many physiological and pathophysiological processes in all organs of the cardiovascular system. From the regulation of blood pressure and blood flow to the adaptive response to end-organ injury, HO plays a critical role in the ability of the cardiovascular system to respond and adapt to changes in homeostasis. There have been great advances in our understanding of the role of HO in the regulation of blood pressure and target organ injury in the last decade. Results from these studies demonstrate that targeting of the HO system could provide novel therapeutic opportunities for the treatment of several cardiovascular and renal diseases. The goal of this review is to highlight the important role of HO in the regulation of cardiovascular and renal function and protection from disease and to highlight areas in which targeting of the HO system needs to be translated to help benefit patient populations.

13.
Hypertension ; 71(6): 1248-1257, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29686012

RESUMO

We determined whether deficiency of neuronal SOCS3 (suppressor of cytokine signaling 3)-a potential negative regulator of leptin signaling-amplifies the chronic effects of leptin on food intake, energy expenditure, glucose, and blood pressure (BP) and protects against adverse cardiometabolic effects of obesity. BP and heart rate were recorded by telemetry, and oxygen consumption (VO2) was monitored in 22-week-old mice with nervous system SOCS3 deficiency (SOCS3-Nestin-Cre) and control mice (SOCS3flox/flox) fed normal or high-fat-high-fructose diet from 6 to 22 weeks of age. Compared with controls, SOCS3-Nestin-Cre mice had lower plasma glucose (124±7 versus 146±10 mg/dL), consumed less food (3.0±0.4 versus 3.6±0.2 g/d), and had similar VO2 (77±6 versus 73±3 mL/kg per minute) and BP (103±3 versus 107±3 mm Hg) but higher heart rate (666±15 versus 602±17 bpm). In mice fed the normal diet, leptin infusion for 7 days caused similar reductions in food intake (2.3±0.1 versus 2.4±0.2 g) but greater increases in BP (15±3 versus 7±2 mm Hg) in SOCS3-Nestin-Cre compared with controls. Leptin reduced blood glucose concentrations in both groups. Male or female SOCS3-Nestin-Cre fed high-fat-high-fructose diet exhibited less weight gain, body fat, and liver steatosis and greater energy expenditure and heart rate compared with controls. Female SOCS3-Nestin-Cre mice fed high-fat-high-fructose diet had higher BP compared with controls. Thus, neuronal SOCS3 seems to play an important role in cardiometabolic regulation because neuronal SOCS3 deficiency reduced body weight and food intake while amplifying leptin's effects on appetite and BP and attenuating the adverse metabolic effects of high-fat-high-fructose diet.


Assuntos
Pressão Sanguínea/fisiologia , Leptina/farmacologia , Síndrome Metabólica/metabolismo , Neurônios/metabolismo , Consumo de Oxigênio/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Animais , Peso Corporal , Modelos Animais de Doenças , Feminino , Masculino , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/fisiopatologia , Camundongos , Transdução de Sinais
14.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R478-R488, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351427

RESUMO

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of leptin receptor signaling and may contribute to leptin resistance in diet-induced obesity. Although PTP1B inhibition has been suggested as a potential weight loss therapy, the role of specific neuronal PTP1B signaling in cardiovascular and metabolic regulation and the importance of sex differences in this regulation are still unclear. In this study, we investigated the impact of proopiomelanocortin (POMC) neuronal PTP1B deficiency in cardiometabolic regulation in male and female mice fed a high-fat diet (HFD). When compared with control mice (PTP1B flox/flox), male and female mice deficient in POMC neuronal PTP1B (PTP1B flox/flox/POMC-Cre) had attenuated body weight gain (males: -18%; females: -16%) and fat mass (males: -33%; female: -29%) in response to HFD. Glucose tolerance was improved by 40%, and liver lipid accumulation was reduced by 40% in PTP1B/POMC-Cre males but not in females. When compared with control mice, deficiency of POMC neuronal PTP1B did not alter mean arterial pressure (MAP) in male or female mice (males: 112 ± 1 vs. 112 ± 1 mmHg in controls; females: 106 ± 3 vs. 109 ± 3 mmHg in controls). Deficiency of POMC neuronal PTP1B also did not alter MAP response to acute stress in males or females compared with control mice (males: Δ32 ± 0 vs. Δ29 ± 4 mmHg; females: Δ22 ± 2 vs. Δ27 ± 4 mmHg). These data demonstrate that POMC-specific PTP1B deficiency improved glucose tolerance and attenuated diet-induced fatty liver only in male mice and attenuated weight gain in males and females but did not enhance the MAP and HR responses to a HFD or to acute stress.


Assuntos
Núcleo Arqueado do Hipotálamo/enzimologia , Glicemia/metabolismo , Intolerância à Glucose/enzimologia , Metabolismo dos Lipídeos , Fígado/metabolismo , Neurônios/enzimologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Obesidade/enzimologia , Pró-Opiomelanocortina/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Núcleo Solitário/enzimologia , Animais , Núcleo Arqueado do Hipotálamo/fisiopatologia , Biomarcadores/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/sangue , Intolerância à Glucose/fisiopatologia , Intolerância à Glucose/prevenção & controle , Fígado/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/etiologia , Obesidade/fisiopatologia , Obesidade/prevenção & controle , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Fatores Sexuais , Núcleo Solitário/fisiopatologia , Aumento de Peso
15.
Physiol Rep ; 5(15)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28784852

RESUMO

Acid-sensing ion channel (ASIC) proteins form extracellular proton-gated, cation-selective channels in neurons and vascular smooth muscle cells and are proposed to act as extracellular proton sensors. However, their importance to vascular responses under conditions associated with extracellular acidosis, such as strenuous exercise, is unclear. Therefore, the purpose of this study was to determine if one ASIC protein, ASIC1a, contributes to extracellular proton-gated vascular responses and exercise tolerance. To determine if ASIC1a contributes to exercise tolerance, we determined peak oxygen (O2) uptake in conscious ASIC1a-/- mice during exhaustive treadmill running. Loss of ASIC1a was associated with a greater peak running speed (60 ± 2 vs. 53 ± 3 m·min-1, P = 0.049) and peak oxygen (O2) uptake during exhaustive treadmill running (9563 ± 120 vs. 8836 ± 276 mL·kg-1·h-1, n = 6-7, P = 0.0082). There were no differences in absolute or relative lean body mass, as determined by EchoMRI. To determine if ASIC1a contributes to vascular responses during muscle contraction, we measured femoral vascular conductance (FVC) during a stepwise electrical stimulation (0.5-5.0 Hz at 3 V for 60 sec) of the left major hind limb muscles. FVC increased to a greater extent in ASIC1a-/- versus ASIC1a+/+ mice (0.44 ± 0.03 vs. 0.30 ± 0.04 mL·min-1·100 g hind limb mass-1 · mmHg-1, n = 5 each, P = 0.0009). Vasodilation following local application of external protons in the spinotrapezius muscle increased the duration, but not the magnitude, of the vasodilatory response in ASIC1a-/- mice. Finally, we examined hind limb vascular density using micro-CT and found increased density of 0-80 µm vessels (P < 0.05). Our findings suggest an increased vascular density and an enhanced vasodilatory response to local protons, to a lesser degree, may contribute to the enhanced vascular conductance and increased peak exercise capacity in ASIC1a-/- mice.


Assuntos
Canais Iônicos Sensíveis a Ácido/fisiologia , Tolerância ao Exercício , Membro Posterior/irrigação sanguínea , Contração Muscular , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/fisiologia , Vasodilatação , Canais Iônicos Sensíveis a Ácido/genética , Animais , Estimulação Elétrica , Feminino , Hiperemia/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Consumo de Oxigênio
16.
Am J Physiol Renal Physiol ; 313(2): F440-F449, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28468964

RESUMO

ANG II has many biological effects in renal physiology, particularly in Ca2+ handling in the regulation of fluid and solute reabsorption. It involves the systemic endocrine renin-angiotensin system (RAS), but tissue and intracrine ANG II are also known. We have shown that ANG II induces heterodimerization of its AT1 and AT2 receptors (AT1R and AT2R) to stimulate sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) activity. Thus, we investigated whether ANG II-AT1R/AT2R complex is formed and internalized, and also examined the intracellular localization of this complex to determine how its effect might be exerted on renal intracrine RAS. Living cell imaging of LLC-PK1 cells, quantification of extracellular ANG II, and use of the receptor antagonists, losartan and PD123319, showed that ANG II is internalized with AT1R/AT2R heterodimers as a complex in a microtubule-dependent and clathrin-independent manner, since colchicine-but not Pitstop2-blocked this process. This result was confirmed by an increase of ß-arrestin phosphorylation after ANG II treatment, clathrin-mediated endocytosis being dependent on dephosphorylation of ß-arrestin. Internalized ANG II colocalized with an endoplasmic reticulum (ER) marker and increased levels of AT1R, AT2R, and PKCα in ER-enriched membrane fractions. This novel evidence suggests the internalization of an ANG II-AT1/AT2 complex to target ER, where it might trigger intracellular Ca2+ responses.


Assuntos
Angiotensina II/metabolismo , Membrana Celular/metabolismo , Endocitose , Retículo Endoplasmático/metabolismo , Rim/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Rim/efeitos dos fármacos , Células LLC-PK1 , Microtúbulos/metabolismo , Complexos Multiproteicos , Fosforilação , Proteína Quinase C-alfa/metabolismo , Transporte Proteico , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Suínos , beta-Arrestinas/metabolismo
17.
J Biol Chem ; 291(48): 25179-25191, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27738106

RESUMO

Non-alcoholic fatty liver disease is the most rapidly growing form of liver disease and if left untreated can result in non-alcoholic steatohepatitis, ultimately resulting in liver cirrhosis and failure. Biliverdin reductase A (BVRA) is a multifunctioning protein primarily responsible for the reduction of biliverdin to bilirubin. Also, BVRA functions as a kinase and transcription factor, regulating several cellular functions. We report here that liver BVRA protects against hepatic steatosis by inhibiting glycogen synthase kinase 3ß (GSK3ß) by enhancing serine 9 phosphorylation, which inhibits its activity. We show that GSK3ß phosphorylates serine 73 (Ser(P)73) of the peroxisome proliferator-activated receptor α (PPARα), which in turn increased ubiquitination and protein turnover, as well as decreased activity. Interestingly, liver-specific BVRA KO mice had increased GSK3ß activity and Ser(P)73 of PPARα, which resulted in decreased PPARα protein and activity. Furthermore, the liver-specific BVRA KO mice exhibited increased plasma glucose and insulin levels and decreased glycogen storage, which may be due to the manifestation of hepatic steatosis observed in the mice. These findings reveal a novel BVRA-GSKß-PPARα axis that regulates hepatic lipid metabolism and may provide unique targets for the treatment of non-alcoholic fatty liver disease.


Assuntos
Metabolismo dos Lipídeos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , PPAR alfa/metabolismo , Proteínas Repressoras/metabolismo , Animais , Glicemia/genética , Glicemia/metabolismo , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , PPAR alfa/genética , Fosforilação , Proteínas Repressoras/genética
18.
Am J Physiol Regul Integr Comp Physiol ; 309(11): R1425-31, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26400187

RESUMO

Cerebrovascular complications and increased risk of encephalopathies are characteristic of preeclampsia and contribute to 40% of preeclampsia/eclampsia-related deaths. Circulating tumor necrosis factor-α (TNF-α) is elevated in preeclamptic women, and infusion of TNF-α into pregnant rats mimics characteristics of preeclampsia. While this suggests that TNF-α has a mechanistic role to promote preeclampsia, the impact of TNF-α on the cerebral vasculature during pregnancy remains unclear. We tested the hypothesis that TNF-α contributes to cerebrovascular abnormalities during placental ischemia by first infusing TNF-α in pregnant rats (200 ng/day ip, from gestational day 14 to 19) at levels to mimic those reported in preeclamptic women. TNF-α increased mean arterial pressure (MAP, P < 0.05) and brain water content in the anterior cerebrum (P < 0.05); however, TNF-α infusion had no effect on blood-brain barrier (BBB) permeability in the anterior cerebrum or posterior cerebrum. We then assessed the role of endogenous TNF-α in mediating these abnormalities in a model of placental ischemia induced by reducing uterine perfusion pressure followed by treatment with the soluble TNF-α receptor (etanercept, 0.8 mg/kg sc) on gestational day 18. Etanercept reduced placental ischemia-mediated increases in MAP, anterior brain water content (P < 0.05), and BBB permeability (202 ± 44% in placental ischemic rats to 101 ± 28% of normal pregnant rats). Our results indicate that TNF-α mechanistically contributes to cerebral edema by increasing BBB permeability and is an underlying factor in the development of cerebrovascular abnormalities associated with preeclampsia complicated by placental ischemia.


Assuntos
Água Corporal/metabolismo , Edema Encefálico/etiologia , Encéfalo/metabolismo , Permeabilidade Capilar , Transtornos Cerebrovasculares/etiologia , Isquemia/complicações , Placenta/irrigação sanguínea , Pré-Eclâmpsia/etiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Pressão Sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Edema Encefálico/prevenção & controle , Permeabilidade Capilar/efeitos dos fármacos , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/fisiopatologia , Transtornos Cerebrovasculares/prevenção & controle , Modelos Animais de Doenças , Etanercepte/administração & dosagem , Feminino , Idade Gestacional , Isquemia/tratamento farmacológico , Isquemia/metabolismo , Isquemia/fisiopatologia , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/fisiopatologia , Pré-Eclâmpsia/prevenção & controle , Gravidez , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/antagonistas & inibidores
20.
J Nephrol Res ; 1(1): 1-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27928552

RESUMO

Pressure-induced constriction (also known as the "myogenic response") is an important mechanodependent response in small renal arteries and arterioles. The response is initiated by vascular smooth muscle cell (VSMC) stretch due to an increase in intraluminal pressure and leads to vasoconstriction. The myogenic response has two important roles as a mechanism of local blood flow autoregulation and protection against systemic blood pressure-induced microvascular damage. However, the molecular mechanisms underlying initiation of myogenic response are unresolved. Although several molecules have been considered initiators of the response, our laboratory has focused on the role of degenerin proteins because of their strong evolutionary link to mechanosensing in the nematode. Our laboratory has addressed the hypothesis that certain degenerin proteins act as mechanosensors in VSMCs. This article discusses the importance of a specific degenerin protein, ß Epithelial Na+ Channel (ßENaC), in pressure-induced vasoconstriction, renal blood flow and susceptibility to renal injury. We propose that loss of the renal myogenic constrictor response delays the correction of renal blood flow that occurs with fluctuations in systemic pressure, which allows pressure swings to be transmitted to the microvasculature, thus increasing the susceptibility to renal injury and hypertension. The role of ßENaC in myogenic regulation is independent of tubular ßENaC and thus represents a non-tubular role for ßENaC in renal-cardiovascular homeostasis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...