Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nature ; 578(7793): E9, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932732

RESUMEN

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nature ; 574(7780): 686-690, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645758

RESUMEN

Dietary habits and vascular risk factors promote both Alzheimer's disease and cognitive impairment caused by vascular factors1-3. Furthermore, accumulation of hyperphosphorylated tau, a microtubule-associated protein and a hallmark of Alzheimer's pathology4, is also linked to vascular cognitive impairment5,6. In mice, a salt-rich diet leads to cognitive dysfunction associated with a nitric oxide deficit in cerebral endothelial cells and cerebral hypoperfusion7. Here we report that dietary salt induces hyperphosphorylation of tau followed by cognitive dysfunction in mice, and that these effects are prevented by restoring endothelial nitric oxide production. The nitric oxide deficiency reduces neuronal calpain nitrosylation and results in enzyme activation, which, in turn, leads to tau phosphorylation by activating cyclin-dependent kinase 5. Salt-induced cognitive impairment is not observed in tau-null mice or in mice treated with anti-tau antibodies, despite persistent cerebral hypoperfusion and neurovascular dysfunction. These findings identify a causal link between dietary salt, endothelial dysfunction and tau pathology, independent of haemodynamic insufficiency. Avoidance of excessive salt intake and maintenance of vascular health may help to stave off the vascular and neurodegenerative pathologies that underlie dementia in the elderly.


Asunto(s)
Disfunción Cognitiva/inducido químicamente , Neuronas/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Humanos , Ratones , Ratones Noqueados , Fosforilación , Cloruro de Sodio Dietético/farmacología
3.
J Cell Physiol ; 239(7): e31360, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38962842

RESUMEN

Junior faculty mentoring committees have important roles in ensuring that faculty thrive and adjust to their new positions and institutions. Here, we describe the purpose, structure, and benefits of junior faculty mentoring committees, which can be a powerful tool for early-career academic investigators in science, technology, engineering, mathematics, and medical (STEMM) fields. There is a paucity of information about what mentoring committees are, how to use them effectively, what areas they should evaluate, and how they can most successfully help junior faculty progress in their careers. This work offers guidance for both junior faculty mentees and mentoring committee members on how to best structure and utilize mentoring committees to promote junior faculty success. A better understanding of the intricacies of the mentoring committee will allow junior faculty members to self-advocate and will equip committee mentors with tools to ensure that junior faculty are successful in thriving in academia.


Asunto(s)
Docentes , Tutoría , Mentores , Humanos , Investigadores/educación
4.
Curr Hypertens Rep ; 26(7): 339-347, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38613621

RESUMEN

PURPOSE OF REVIEW: Cardiovascular disease (CVD) is a leading cause of death and chronic disability worldwide. Yet, despite extensive intervention strategies the number of persons affected by CVD continues to rise. Thus, there is great interest in unveiling novel mechanisms that may lead to new treatments. Considering this dilemma, recent focus has turned to the neuroimmune mechanisms involved in CVD pathology leading to a deeper understanding of the brain's involvement in disease pathology. This review provides an overview of new and salient findings regarding the neuroimmune mechanisms that contribute to CVD. RECENT FINDINGS: The brain contains neuroimmune niches comprised of glia in the parenchyma and immune cells at the brain's borders, and there is strong evidence that these neuroimmune niches are important in both health and disease. Mechanistic studies suggest that the activation of glia and immune cells in these niches modulates CVD progression in hypertension and heart failure and contributes to the inevitable end-organ damage to the brain. This review provides evidence supporting the role of neuroimmune niches in CVD progression. However, additional research is needed to understand the effects of prolonged neuroimmune activation on brain function.


Asunto(s)
Encéfalo , Enfermedades Cardiovasculares , Neuroinmunomodulación , Humanos , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/fisiopatología , Neuroinmunomodulación/fisiología , Encéfalo/inmunología , Encéfalo/fisiopatología , Encéfalo/patología , Neuroglía/inmunología , Animales
5.
Am J Physiol Heart Circ Physiol ; 317(2): H279-H289, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31150271

RESUMEN

Increased sympathetic nervous system activity is a hallmark of hypertension (HTN), and it is implicated in altered immune system responses in its pathophysiology. However, the precise mechanisms of neural-immune interaction in HTN remain elusive. We have previously shown an association between elevated sympathetic drive to the bone marrow (BM) and activated BM immune cells in rodent models of HTN. Moreover, microglial-dependent neuroinflammation is also seen in rodent models of HTN. However, the cause-effect relationship between central and systemic inflammatory responses and the sympathetic drive remains unknown. These observations led us to hypothesize that increase in the femoral BM sympathetic nerve activity (fSNA) initiates a cascade of events leading to increase in blood pressure (BP). Here, we investigated the temporal relationship between the BM sympathetic drive, activation of the central and peripheral immune system, and increase in BP in the events leading to established HTN. The present study demonstrates that central infusion of angiotensin II (ANG II) induces early microglial activation in the paraventricular nucleus of hypothalamus, which preceded increase in the fSNA. In turn, activation of fSNA correlated with the timing of increased production and release of CD4+.IL17+ T cells and other proinflammatory cells into circulation and elevation in BP, whereas infiltration of CD4+ cells to the paraventricular nucleus marked establishment of ANG II HTN. This study identifies cellular and molecular mechanisms involved in neural-immune interactions in early and established stages of rodent ANG II HTN. NEW & NOTEWORTHY Early microglia activation in paraventricular nucleus precedes sympathetic activation of the bone marrow. This leads to increased bone marrow immune cells and their release into circulation and an increase in blood pressure. Infiltration of CD4+ T cells into paraventricular nucleus paraventricular nucleus marks late hypertension.


Asunto(s)
Presión Sanguínea , Médula Ósea/inervación , Hipertensión/fisiopatología , Inflamación/fisiopatología , Neuroinmunomodulación , Núcleo Hipotalámico Paraventricular/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Angiotensina II , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Fémur , Hipertensión/inducido químicamente , Hipertensión/inmunología , Hipertensión/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Microglía/inmunología , Microglía/metabolismo , Núcleo Hipotalámico Paraventricular/inmunología , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Sprague-Dawley , Sistema Nervioso Simpático/inmunología , Sistema Nervioso Simpático/metabolismo , Factores de Tiempo
6.
Circ Res ; 120(2): 312-323, 2017 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-27799253

RESUMEN

RATIONALE: Sympathetic nervous system control of inflammation plays a central role in hypertension. The gut receives significant sympathetic innervation, is densely populated with a diverse microbial ecosystem, and contains immune cells that greatly impact overall inflammatory homeostasis. Despite this uniqueness, little is known about the involvement of the gut in hypertension. OBJECTIVE: Test the hypothesis that increased sympathetic drive to the gut is associated with increased gut wall permeability, increased inflammatory status, and microbial dysbiosis and that these gut pathological changes are linked to hypertension. METHODS AND RESULTS: Gut epithelial integrity and wall pathology were examined in spontaneously hypertensive rat and chronic angiotensin II infusion rat models. The increase in blood pressure in spontaneously hypertensive rat was associated with gut pathology that included increased intestinal permeability and decreased tight junction proteins. These changes in gut pathology in hypertension were associated with alterations in microbial communities relevant in blood pressure control. We also observed enhanced gut-neuronal communication in hypertension originating from paraventricular nucleus of the hypothalamus and presenting as increased sympathetic drive to the gut. Finally, angiotensin-converting enzyme inhibition (captopril) normalized blood pressure and was associated with reversal of gut pathology. CONCLUSIONS: A dysfunctional sympathetic-gut communication is associated with gut pathology, dysbiosis, and inflammation and plays a key role in hypertension. Thus, targeting of gut microbiota by innovative probiotics, antibiotics, and fecal transplant, in combination with the current pharmacotherapy, may be a novel strategy for hypertension treatment.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatología , Angiotensina II/toxicidad , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Masculino , Permeabilidad/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Ratas Wistar
7.
Circ Res ; 118(8): 1327-36, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27081113

RESUMEN

Hypertension is the most prevalent modifiable risk factor for cardiovascular disease and disorders directly influencing cardiovascular disease morbidity and mortality, such as diabetes mellitus, chronic kidney disease, obstructive sleep apnea, etc. Despite aggressive attempts to influence lifestyle modifications and advances in pharmacotherapeutics, a large percentage of patients still do not achieve recommended blood pressure control worldwide. Thus, we think that mechanism-based novel strategies should be considered to significantly improve control and management of hypertension. The overall objective of this review is to summarize implications of peripheral- and neuroinflammation as well as the autonomic nervous system-bone marrow communication in hematopoietic cell homeostasis and their impact on hypertension pathophysiology. In addition, we discuss the novel and emerging field of intestinal microbiota and roles of gut permeability and dysbiosis in cardiovascular disease and hypertension. Finally, we propose a brain-gut-bone marrow triangular interaction hypothesis and discuss its potential in the development of novel therapies for hypertension.


Asunto(s)
Médula Ósea/metabolismo , Encéfalo/metabolismo , Sistema Nervioso Entérico/metabolismo , Tracto Gastrointestinal/metabolismo , Hipertensión/metabolismo , Hipertensión/terapia , Animales , Microbioma Gastrointestinal/fisiología , Humanos , Hipertensión/diagnóstico , Conducta de Reducción del Riesgo
8.
Circ Res ; 117(2): 178-91, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25963715

RESUMEN

RATIONALE: Microglial activation in autonomic brain regions is a hallmark of neuroinflammation in neurogenic hypertension. Despite evidence that an impaired sympathetic nerve activity supplying the bone marrow (BM) increases inflammatory cells and decreases angiogenic cells, little is known about the reciprocal impact of BM-derived inflammatory cells on neuroinflammation in hypertension. OBJECTIVE: To test the hypothesis that proinflammatory BM cells from hypertensive animals contribute to neuroinflammation and hypertension via a brain-BM interaction. METHODS AND RESULTS: After BM ablation in spontaneously hypertensive rats, and reconstitution with normotensive Wistar Kyoto rat BM, the resultant chimeric spontaneously hypertensive rats displayed significant reduction in mean arterial pressure associated with attenuation of both central and peripheral inflammation. In contrast, an elevated mean arterial pressure along with increased central and peripheral inflammation was observed in chimeric Wistar-Kyoto rats reconstituted with spontaneously hypertensive rat BM. Oral treatment with minocycline, an inhibitor of microglial activation, attenuated hypertension in both the spontaneously hypertensive rats and the chronic angiotensin II-infused rats. This was accompanied by decreased sympathetic drive and inflammation. Furthermore, in chronic angiotensin II-infused rats, minocycline prevented extravasation of BM-derived cells to the hypothalamic paraventricular nucleus, presumably via a mechanism of decreased C-C chemokine ligand 2 levels in the cerebrospinal fluid. CONCLUSIONS: The BM contributes to hypertension by increasing peripheral inflammatory cells and their extravasation into the brain. Minocycline is an effective therapy to modify neurogenic components of hypertension. These observations support the hypothesis that BM-derived cells are involved in neuroinflammation, and targeting them may be an innovative strategy for neurogenic resistant hypertension therapy.


Asunto(s)
Células de la Médula Ósea/fisiología , Hipertensión/etiología , Microglía/fisiología , Inflamación Neurogénica/complicaciones , Núcleo Hipotalámico Paraventricular/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Angiotensina II , Animales , Barorreflejo/fisiología , Trasplante de Médula Ósea , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Femenino , Hipertensión/fisiopatología , Hipertensión/prevención & control , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Masculino , Microglía/efectos de los fármacos , Minociclina/uso terapéutico , Norepinefrina/sangre , Núcleo Hipotalámico Paraventricular/inmunología , Quimera por Radiación , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Sistema Nervioso Simpático/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
9.
Aging Cell ; 23(2): e14042, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38030595

RESUMEN

The article "Neuronal induction of BNIP3-mediated mitophagy slows systemic aging in Drosophila" reveals BCL2-interacting protein 3 as a therapeutic target to counteract brain aging and prolong overall organismal health with age. In this spotlight, we consider the roles of BNIP3, a mitochondrial outer membrane protein, in the adult nervous system, including its induction of mitophagy and prevention of dysfunctional mitochondria in the aged brain. Implications for other tissue types to reduce the burden of aging are further considered.


Asunto(s)
Mitocondrias , Proteínas Mitocondriales , Proteínas Mitocondriales/metabolismo , Mitocondrias/metabolismo , Mitofagia , Encéfalo/metabolismo
10.
Hypertension ; 81(1): 34-44, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37732479

RESUMEN

Dementia affects almost 50 million adults worldwide, and remains a major cause of death and disability. Hypertension is a leading risk factor for dementia, including Alzheimer disease and Alzheimer disease-related dementias. Although this association is well-established, the mechanisms underlying hypertension-induced cognitive decline remain poorly understood. By exploring the mechanisms mediating the detrimental effects of hypertension on the brain, studies have aimed to provide therapeutic insights and strategies on how to protect the brain from the effects of blood pressure elevation. In this review, we focus on the basic mechanisms contributing to the cerebrovascular adaptions to elevated blood pressure and hypertension-induced microvascular injury. We also assess the cellular mechanisms of neurovascular unit dysfunction, focusing on the premise that cognitive impairment ensues when the dynamic metabolic demands of neurons are not met due to neurovascular uncoupling, and summarize cognitive deficits across various rodent models of hypertension as a resource for investigators. Despite significant advances in antihypertensive therapy, hypertension remains a critical risk factor for cognitive decline, and several questions remain about the development and progression of hypertension-induced cognitive impairment.


Asunto(s)
Enfermedad de Alzheimer , Lesiones Encefálicas , Disfunción Cognitiva , Hipertensión , Humanos , Disfunción Cognitiva/etiología , Encéfalo/metabolismo
11.
Nat Neurosci ; 27(1): 63-77, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38049579

RESUMEN

Hypertension (HTN), a disease afflicting over one billion individuals worldwide, is a leading cause of cognitive impairment, the mechanisms of which remain poorly understood. In the present study, in a mouse model of HTN, we find that the neurovascular and cognitive dysfunction depends on interleukin (IL)-17, a cytokine elevated in individuals with HTN. However, neither circulating IL-17 nor brain angiotensin signaling can account for the dysfunction. Rather, IL-17 produced by T cells in the dura mater is the mediator released in the cerebrospinal fluid and activating IL-17 receptors on border-associated macrophages (BAMs). Accordingly, depleting BAMs, deleting IL-17 receptor A in brain macrophages or suppressing meningeal T cells rescues cognitive function without attenuating blood pressure elevation, circulating IL-17 or brain angiotensin signaling. Our data unveil a critical role of meningeal T cells and macrophage IL-17 signaling in the neurovascular and cognitive dysfunction in a mouse model of HTN.


Asunto(s)
Disfunción Cognitiva , Hipertensión , Ratones , Animales , Interleucina-17 , Angiotensina II , Linfocitos T , Cloruro de Sodio Dietético
12.
Curr Hypertens Rep ; 15(4): 377-89, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23715920

RESUMEN

It is widely accepted that the pathophysiology of hypertension involves autonomic nervous system dysfunction, as well as a multitude of immune responses. However, the close interplay of these systems in the development and establishment of high blood pressure and its associated pathophysiology remains elusive and is the subject of extensive investigation. It has been proposed that an imbalance of the neuro-immune systems is a result of an enhancement of the "proinflammatory sympathetic" arm in conjunction with dampening of the "anti-inflammatory parasympathetic" arm of the autonomic nervous system. In addition to the neuronal modulation of the immune system, it is proposed that key inflammatory responses are relayed back to the central nervous system and alter the neuronal communication to the periphery. The overall objective of this review is to critically discuss recent advances in the understanding of autonomic immune modulation, and propose a unifying hypothesis underlying the mechanisms leading to the development and maintenance of hypertension, with particular emphasis on the bone marrow, as it is a crucial meeting point for neural, immune, and vascular networks.


Asunto(s)
Médula Ósea/fisiopatología , Encéfalo/fisiopatología , Hipertensión/fisiopatología , Animales , Encéfalo/inmunología , Humanos , Inflamación/fisiopatología , Neuroinmunomodulación/inmunología , Neuroinmunomodulación/fisiología , Sistema Nervioso Simpático/fisiopatología
13.
Hypertension ; 80(1): 22-34, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36129176

RESUMEN

Hypertension affects a significant proportion of the adult and aging population and represents an important risk factor for vascular cognitive impairment and late-life dementia. Chronic high blood pressure continuously challenges the structural and functional integrity of the cerebral vasculature, leading to microvascular rarefaction and dysfunction, and neurovascular uncoupling that typically impairs cerebral blood supply. Hypertension disrupts blood-brain barrier integrity, promotes neuroinflammation, and may contribute to amyloid deposition and Alzheimer pathology. The mechanisms underlying these harmful effects are still a focus of investigation, but studies in animal models have provided significant molecular and cellular mechanistic insights. Remaining questions relate to whether adequate treatment of hypertension may prevent deterioration of cognitive function, the threshold for blood pressure treatment, and the most effective antihypertensive drugs. Recent advances in neurovascular biology, advanced brain imaging, and detection of subtle behavioral phenotypes have begun to provide insights into these critical issues. Importantly, a parallel analysis of these parameters in animal models and humans is feasible, making it possible to foster translational advancements. In this review, we provide a critical evaluation of the evidence available in experimental models and humans to examine the progress made and identify remaining gaps in knowledge.


Asunto(s)
Disfunción Cognitiva , Hipertensión , Humanos , Anciano , Disfunción Cognitiva/etiología , Hipertensión/tratamiento farmacológico
14.
Hypertension ; 76(3): 795-807, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32654560

RESUMEN

Hypertension is a leading cause of stroke and dementia, effects attributed to disrupting delivery of blood flow to the brain. Hypertension also alters the blood-brain barrier (BBB), a critical component of brain health. Although endothelial cells are ultimately responsible for the BBB, the development and maintenance of the barrier properties depend on the interaction with other vascular-associated cells. However, it remains unclear if BBB disruption in hypertension requires cooperative interaction with other cells. Perivascular macrophages (PVM), innate immune cells closely associated with cerebral microvessels, have emerged as major contributors to neurovascular dysfunction. Using 2-photon microscopy in vivo and electron microscopy in a mouse model of Ang II (angiotensin II) hypertension, we found that the vascular segments most susceptible to increased BBB permeability are arterioles and venules >10 µm and not capillaries. Brain macrophage depletion with clodronate attenuates, but does not abolish, the increased BBB permeability in these arterioles where PVM are located. Deletion of AT1R (Ang II type-1 receptors) in PVM using bone marrow chimeras partially attenuated the BBB dysfunction through the free radical-producing enzyme Nox2. In contrast, downregulation of AT1R in cerebral endothelial cells using a viral gene transfer-based approach prevented the BBB disruption completely. The results indicate that while endothelial AT1R, mainly in arterioles and venules, initiate the BBB disruption in hypertension, PVM are required for the full expression of the dysfunction. The findings unveil a previously unappreciated contribution of resident brain macrophages to increased BBB permeability of hypertension and identify PVM as a putative therapeutic target in diseases associated with BBB dysfunction.


Asunto(s)
Arteriolas/fisiopatología , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Circulación Cerebrovascular/fisiología , Endotelio Vascular , Hipertensión , Macrófagos/fisiología , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Permeabilidad Capilar/fisiología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Sistema Glinfático/inmunología , Sistema Glinfático/patología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Ratones
15.
J Cereb Blood Flow Metab ; 38(12): 2112-2128, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30295560

RESUMEN

Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.


Asunto(s)
Disfunción Cognitiva/complicaciones , Hipertensión/complicaciones , Cloruro de Sodio Dietético/efectos adversos , Animales , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/fisiopatología , Disfunción Cognitiva/fisiopatología , Humanos , Hipertensión/fisiopatología , Acoplamiento Neurovascular
16.
Nat Neurosci ; 21(2): 240-249, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29335605

RESUMEN

A diet rich in salt is linked to an increased risk of cerebrovascular diseases and dementia, but it remains unclear how dietary salt harms the brain. We report that, in mice, excess dietary salt suppresses resting cerebral blood flow and endothelial function, leading to cognitive impairment. The effect depends on expansion of TH17 cells in the small intestine, resulting in a marked increase in plasma interleukin-17 (IL-17). Circulating IL-17, in turn, promotes endothelial dysfunction and cognitive impairment by the Rho kinase-dependent inhibitory phosphorylation of endothelial nitric oxide synthase and reduced nitric oxide production in cerebral endothelial cells. The findings reveal a new gut-brain axis linking dietary habits to cognitive impairment through a gut-initiated adaptive immune response compromising brain function via circulating IL-17. Thus, the TH17 cell-IL-17 pathway is a putative target to counter the deleterious brain effects induced by dietary salt and other diseases associated with TH17 polarization.


Asunto(s)
Trastornos Cerebrovasculares/inducido químicamente , Trastornos del Conocimiento/inducido químicamente , Intestino Delgado/patología , Cloruro de Sodio Dietético/toxicidad , Células Th17/efectos de los fármacos , Acetilcolina/farmacología , Amidas/farmacología , Animales , Antihipertensivos/farmacología , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Trastornos Cerebrovasculares/tratamiento farmacológico , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interleucina-17/administración & dosificación , Interleucina-17/sangre , Interleucina-17/genética , Interleucina-17/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Acoplamiento Neurovascular/efectos de los fármacos , Fosforilación/efectos de los fármacos , Piridinas/farmacología
17.
Front Physiol ; 8: 592, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28912720

RESUMEN

Activation of autonomic neural pathways by chronic hypertensive stimuli plays a significant role in pathogenesis of hypertension. Here, we proposed that even a single acute hypertensive stimulus will activate neural and immune pathways that may be important in initiation of memory imprinting seen in chronic hypertension. We investigated the effects of acute angiotensin II (Ang II) administration on blood pressure, neural activation in cardioregulatory brain regions, and central and systemic immune responses, at 1 and 24 h post-injection. Administration of a single bolus intra-peritoneal (I.P.) injection of Ang II (36 µg/kg) resulted in a transient increase in the mean arterial pressure (MAP) (by 22 ± 4 mmHg vs saline), which returned to baseline within 1 h. However, in contrast to MAP, neuronal activity, as measured by manganese-enhanced magnetic resonance (MEMRI), remained elevated in several cardioregulatory brain regions over 24 h. The increase was predominant in autonomic regions, such as the subfornical organ (SFO; ~20%), paraventricular nucleus of the hypothalamus (PVN; ~20%) and rostral ventrolateral medulla (RVLM; ~900%), among others. Similarly, systemic and central immune responses, as evidenced by circulating levels of CD4+/IL17+ T cells, and increased IL17 levels and activation of microglia in the PVN, respectively, remained elevated at 24 h following Ang II challenge. Elevated Fos expression in the PVN was also present at 24 h (by 73 ± 11%) following Ang II compared to control saline injections, confirming persistent activation of PVN. Thus, even a single Ang II hypertensive stimulus will initiate changes in neuronal and immune cells that play a role in the developing hypertensive phenotype.

19.
J Clin Invest ; 126(12): 4674-4689, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27841763

RESUMEN

Hypertension is a leading risk factor for dementia, but the mechanisms underlying its damaging effects on the brain are poorly understood. Due to a lack of energy reserves, the brain relies on continuous delivery of blood flow to its active regions in accordance with their dynamic metabolic needs. Hypertension disrupts these vital regulatory mechanisms, leading to the neuronal dysfunction and damage underlying cognitive impairment. Elucidating the cellular bases of these impairments is essential for developing new therapies. Perivascular macrophages (PVMs) represent a distinct population of resident brain macrophages that serves key homeostatic roles but also has the potential to generate large amounts of reactive oxygen species (ROS). Here, we report that PVMs are critical in driving the alterations in neurovascular regulation and attendant cognitive impairment in mouse models of hypertension. This effect was mediated by an increase in blood-brain barrier permeability that allowed angiotensin II to enter the perivascular space and activate angiotensin type 1 receptors in PVMs, leading to production of ROS through the superoxide-producing enzyme NOX2. These findings unveil a pathogenic role of PVMs in the neurovascular and cognitive dysfunction associated with hypertension and identify these cells as a putative therapeutic target for diseases associated with cerebrovascular oxidative stress.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Disfunción Cognitiva/metabolismo , Hipertensión/metabolismo , Macrófagos/metabolismo , Estrés Oxidativo , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Barrera Hematoencefálica/patología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Hipertensión/complicaciones , Hipertensión/genética , Hipertensión/patología , Macrófagos/patología , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo
20.
Acta Biomater ; 45: 296-302, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27567964

RESUMEN

Hypertension is the most prevalent risk factor for cardiovascular disease caused by a persistent increase in arterial blood pressure that has lasting effects on the mechanical properties of affected tissues like myocardium and blood vessels. Our group recently discovered that gut dysbiosis is linked to hypertension in several animal models and humans; however, whether hypertension influences the gut's mechanical properties remains unknown. In this study, we evaluated the hypothesis that hypertension increases fibrosis and thus mechanical properties of the gut. A custom indentation system was used to test colon samples from Wistar Kyoto (WKY) normotensive rats and Spontaneously Hypertensive Rats (SHR). Using force-displacement data, we derived an steady-state modulus metric to quantify mechanical properties of gastrointestinal tissue. We observed that SHR proximal colon has a mean steady-state modulus almost 3 times greater than WKY control rat colon (5.11±1.58kPa and 18.17±11.45kPa, respectively). These increases were associated with increase in vascular smooth muscle cells layer and collagen deposition in the intestinal wall in the SHR. STATEMENT OF SIGNIFICANCE: Mechanical characterization of biological materials can provide insight into health and disease of tissue. Recent investigations into a variety of cardiovascular pathologies show coincident changes in the microbiome and pathology of the gut. In this study, we sought to quantify changes in the gut in hypertension through mechanical characterization. Our methods and simple models for characterization, adapted from Hertz indentation models, prove useful to identify a meaningful steady-state modulus metric for small and irregular tissues from laboratory animals. Our data, for the first time, establish a stiffening of the gut wall in Spontaneously Hypertensive Rats. This observation suggests significant structural and functional changes in the gut correlate with hypertension, and future experiments are warranted to explore the specific causal relationship between dysbiosis, fibrosis, and stiffening in the gut during the development and maintenance of hypertension.


Asunto(s)
Tracto Gastrointestinal/fisiopatología , Hipertensión/fisiopatología , Animales , Fenómenos Biomecánicos , Colágeno/metabolismo , Módulo de Elasticidad , Músculo Liso/fisiopatología , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA