Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 326(5): R416-R426, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38406845

ABSTRACT

Cerebrovascular reactivity (CVR) decreases with advancing age, contributing to increased risk of cognitive impairment; however, the mechanisms underlying the age-related decrease in CVR are incompletely understood. Age-related changes to T cells, such as impaired mitochondrial respiration, increased inflammation, likely contribute to peripheral and cerebrovascular dysfunction in animals. However, whether T-cell mitochondrial respiration is related to cerebrovascular function in humans is not known. Therefore, we hypothesized that peripheral T-cell mitochondrial respiration would be positively associated with CVR and that T-cell glycolytic metabolism would be negatively associated with CVR. Twenty middle-aged adults (58 ± 5 yr) were recruited for this study. T cells were separated from peripheral blood mononuclear cells. Cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR, a marker of glycolytic activity) were measured using extracellular flux analysis. CVR was quantified using the breath-hold index (BHI), which reflects the change in blood velocity in the middle-cerebral artery (MCAv) during a 30-s breath-hold. In contrast to our hypothesis, we found that basal OCR in CD8+ T cells (ß = -0.59, R2 = 0.27, P = 0.019) was negatively associated with BHI. However, in accordance with our hypothesis, we found that basal ECAR (ß = -2.20, R2 = 0.29, P = 0.015) and maximum ECAR (ß = -50, R2 = 0.24, P = 0.029) were negatively associated with BHI in CD8+ T cells. There were no associations observed in CD4+ T cells. These associations appeared to be primarily mediated by an association with the pressor response to the breath-hold test. Overall, our findings suggest that CD8+ T-cell respiration and glycolytic activity may influence CVR in humans.NEW & NOTEWORTHY Peripheral T-cell metabolism is related to in vivo cerebrovascular reactivity in humans. Higher glycolytic metabolism in CD8+ T cells was associated with lower cerebrovascular reactivity to a breath-hold in middle-aged adults, which is possibly reflective of a more proinflammatory state in midlife.


Subject(s)
CD8-Positive T-Lymphocytes , Leukocytes, Mononuclear , Adult , Humans , Middle Aged , Cerebrovascular Circulation/physiology , Respiration , Breath Holding
2.
Hypertension ; 81(3): 530-540, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38193292

ABSTRACT

BACKGROUND: CD8+ T cells (CD8Ts) have been implicated in hypertension. However, the specific mechanisms are not fully understood. In this study, we explore the contribution of the P2X7 (purinergic receptor P2X7) receptor to CD8T activation and subsequent promotion of sodium retention in the kidney. METHODS: We used mouse models of hypertension. Wild type were used as genetic controls, OT1 and Rag2/OT1 mice were utilized to determine antigen dependency, and P2X7-knockout mice were studied to define the role of P2X7 in activating CD8Ts and promoting hypertension. Blood pressure was monitored continuously and kidneys were obtained at different experimental end points. Freshly isolated CD8Ts from mice for activation assays and ATP stimulation. CD8T activation-induced promotion of sodium retention was explored in cocultures of CD8Ts and mouse DCTs. RESULTS: We found that OT1 and Rag2/OT1 mice, which are nonresponsive to common antigens, still developed hypertension and CD8T-activation in response to deoxycorticosterone acetate/salt treatment, similar to wild-type mice. Further studies identified the P2X7 receptor on CD8Ts as a possible mediator of this antigen-independent activation of CD8Ts in hypertension. Knockout of the P2X7 receptor prevented calcium influx and cytokine production in CD8Ts. This finding was associated with reduced CD8T-DCT stimulation, reversal of excessive salt retention in DCTs, and attenuated development of salt-sensitive hypertension. CONCLUSIONS: Our findings suggest a novel mechanism by which CD8Ts are activated in hypertension to exacerbate salt retention and infer that the P2X7 receptor on CD8Ts may represent a new therapeutic target to attenuate T-cell-mediated immunopathology in hypertension.


Subject(s)
CD8-Positive T-Lymphocytes , Hypertension , Mice , Animals , Receptors, Purinergic P2X7/genetics , Mice, Knockout , Sodium Chloride, Dietary , Sodium , Adenosine Triphosphate , Mice, Inbred C57BL
3.
Geroscience ; 46(1): 1035-1051, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37354388

ABSTRACT

Aging of the arteries is characterized by increased large artery stiffness and impaired endothelium-dependent dilation. We have previously shown that in old (22-24 month) mice T cells accumulate within aorta and mesentery. We have also shown that pharmacologic and genetic deletion of these T cells ameliorates age-related arterial dysfunction. These data indicate that T cells contribute to arterial aging; however, it is unknown if aged T cells alone can induce arterial dysfunction in otherwise young mice. To produce an aged-like T cell phenotype, mice were thymectomized at three-weeks of age or were left with their thymus intact. At 9 months of age, thymectomized mice exhibited greater proportions of both CD4 + and CD8 + memory T cells compared to controls in the blood. Similar changes were observed in the T cells accumulating in the aorta and mesentery. We also observed greater numbers of proinflammatory cytokine producing T cells in the aorta and mesentery. The phenotypic T cell changes in the blood, aorta and mesentery of thymectomized mice were similar to those observed when we compared young (4-6 month) to old thymus intact mice. Along with these alterations, compared to controls, thymectomized mice exhibited augmented large artery stiffness and greater aortic collagen deposition as well as impaired mesenteric artery endothelium dependent dilation due to blunted nitric oxide bioavailability. These results indicate that early life thymectomy results in arterial dysfunction and suggest that an aged-like T cell phenotype alone is sufficient to induce arterial dysfunction in otherwise young mice.


Subject(s)
Arteries , Thymectomy , Animals , Mice , Aging , Thymus Gland , Aorta
4.
PLoS One ; 18(5): e0285253, 2023.
Article in English | MEDLINE | ID: mdl-37163513

ABSTRACT

Atherosclerosis is the root cause of major cardiovascular diseases (CVD) such as myocardial infarction and stroke. ADP-ribosylation factor 6 (Arf6) is a ubiquitously expressed GTPase known to be involved in inflammation, vascular permeability and is sensitive to changes in shear stress. Here, using atheroprone, ApoE-/- mice, with a single allele deletion of Arf6 (HET) or wildtype Arf6 (WT), we demonstrate that reduction in Arf6 attenuates atherosclerotic plaque burden and severity. We found that plaque burden in the descending aorta was lower in HET compared to WT mice (p˂0.001) after the consumption of an atherogenic Paigen diet for 5 weeks. Likewise, luminal occlusion, necrotic core size, plaque grade, elastic lamina breaks, and matrix deposition were lower in the aortic root atheromas of HET compared to WT mice (all p≤0.05). We also induced advanced human-like complex atherosclerotic plaque in the left carotid artery using partial carotid ligation surgery and found that atheroma area, plaque grade, intimal necrosis, intraplaque hemorrhage, thrombosis, and calcification were lower in HET compared to WT mice (all p≤0.04). Our findings suggest that the atheroprotection afforded by Arf6 heterozygosity may result from reduced immune cell migration (all p≤0.005) as well as endothelial and vascular smooth muscle cell proliferation (both p≤0.001) but independent of changes in circulating lipids (all p≥0.40). These findings demonstrate a critical role for Arf6 in the development and severity of atherosclerosis and suggest that Arf6 inhibition can be explored as a novel therapeutic strategy for the treatment of atherosclerotic CVD.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Humans , Mice , ADP-Ribosylation Factor 6 , Aorta , Atherosclerosis/genetics , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Plaque, Atherosclerotic/genetics
5.
Geroscience ; 45(4): 2351-2365, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36787090

ABSTRACT

Advanced age is accompanied by arterial dysfunction, as well as a diminished glycocalyx, which may be linked to reduced high molecular weight-hyaluronan (HMW-HA) synthesis. However, the impact of glycocalyx deterioration in age-related arterial dysfunction is unknown. We sought to determine if manipulations in glycocalyx properties would alter arterial function. Tamoxifen-induced hyaluronan synthase 2 (Has2) reduction was used to decrease glycocalyx properties. Three weeks post-tamoxifen treatment, glycocalyx thickness was lower in Has2 knockout compared to wild-type mice (P<0.05). Has2 reduction induced arterial dysfunction, demonstrated by impaired endothelium-dependent dilation (EDD) and elevated aortic stiffness (P<0.05). To augment glycocalyx properties, old mice received 10 weeks of a glycocalyx-targeted therapy via Endocalyx™ (old+ECX), which contains HMW-HA and other glycocalyx components. Compared to old control mice, glycocalyx properties and EDD were augmented, and aortic stiffness decreased in old+ECX mice (P<0.05). Old+ECX mice had a more youthful aortic phenotype, demonstrated by lower collagen content and higher elastin content than old control mice (P<0.05). Functional outcomes were repeated in old mice that underwent a diet supplemented solely with HMW-HA (old+HA). Compared to old controls, glycocalyx properties and EDD were augmented, and aortic stiffness was lower in old+HA mice (P<0.05). We did not observe any differences between old+HA and old+ECX mice (P>0.05). Has2 reduction phenocopies age-related arterial dysfunction, while 10 weeks of glycocalyx-targeted therapy that restores the glycocalyx also ameliorates age-related arterial dysfunction. These findings suggest that the glycocalyx may be a viable therapeutic target to ameliorate age-related arterial dysfunction.


Subject(s)
Arteries , Glycocalyx , Animals , Mice , Aorta , Dietary Supplements , Tamoxifen
6.
Aging Cell ; 22(2): e13767, 2023 02.
Article in English | MEDLINE | ID: mdl-36637079

ABSTRACT

Aging results in an elevated burden of senescent cells, senescence-associated secretory phenotype (SASP), and tissue infiltration of immune cells contributing to chronic low-grade inflammation and a host of age-related diseases. Recent evidence suggests that the clearance of senescent cells alleviates chronic inflammation and its associated dysfunction and diseases. However, the effect of this intervention on metabolic function in old age remains poorly understood. Here, we demonstrate that dasatinib and quercetin (D&Q) have senolytic effects, reducing age-related increase in senescence-associated ß-galactosidase, expression of p16 and p21 gene and P16 protein in perigonadal white adipose tissue (pgWAT; all p ≤ 0.04). This treatment also suppressed age-related increase in the expression of a subset of pro-inflammatory SASP genes (mcp1, tnf-α, il-1α, il-1ß, il-6, cxcl2, and cxcl10), crown-like structures, abundance of T cells and macrophages in pgWAT (all p ≤ 0.04). In the liver and skeletal muscle, we did not find a robust effect of D&Q on senescence and inflammatory SASP markers. Although we did not observe an age-related difference in glucose tolerance, D&Q treatment improved fasting blood glucose (p = 0.001) and glucose tolerance (p = 0.007) in old mice that was concomitant with lower hepatic gluconeogenesis. Additionally, D&Q improved insulin-stimulated suppression of plasma NEFAs (p = 0.01), reduced fed and fasted plasma triglycerides (both p ≤ 0.04), and improved systemic lipid tolerance (p = 0.006). Collectively, results from this study suggest that D&Q attenuates adipose tissue inflammation and improves systemic metabolic function in old age. These findings have implications for the development of therapeutic agents to combat metabolic dysfunction and diseases in old age.


Subject(s)
Cellular Senescence , Quercetin , Mice , Animals , Dasatinib/pharmacology , Dasatinib/therapeutic use , Cellular Senescence/genetics , Quercetin/pharmacology , Quercetin/therapeutic use , Senotherapeutics , Adipose Tissue/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Glucose/metabolism
7.
J Physiol ; 601(3): 391-392, 2023 02.
Article in English | MEDLINE | ID: mdl-36625161
8.
Am J Physiol Heart Circ Physiol ; 322(4): H568-H574, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35179977

ABSTRACT

The prevalence of major depressive disorder (MDD) is highest in young adulthood, an effect that has been magnified by the COVID-19 pandemic. Importantly, individuals with MDD are at a greater risk of developing cardiovascular disease (CVD). Accumulating evidence supports immune system dysregulation as a major contributor to the elevated CVD risk in older adults with MDD; however, whether this is present in young adults with MDD without comorbid disease remains unclear. Interestingly, recent data suggest augmented T-cell mitochondrial reactive oxygen species (T-cell mitoROS) as a potent driver of immune dysregulation in animal models of psychiatric disease. With this background in mind, we tested the hypothesis that young adults with MDD would have augmented T-cell mitoROS and circulating proinflammatory cytokines compared with healthy young adults without MDD (HA). Whole blood was drawn from 14 young adults with MDD (age: 23 ± 2 yr) and 11 HA (age: 22 ± 1 yr). T-cell mitoROS (MitoSOX red; total: CD3+, T-helper: CD4+, T cytotoxic: CD8+) and serum cytokines were assessed by flow cytometry. Total T-cell mitoROS was significantly greater in adults with MDD compared with HA [median: 14,089 arbitrary units (AU); median: 1,362 AU, P = 0.01]. Likewise, both T-helper and T-cytotoxic cell mitoROS were significantly greater in adults with MDD compared with HA (both: P < 0.05). There were no differences in circulating cytokines between groups (all cytokines: P > 0.05). Collectively, these findings suggest that elevated T-cell mitoROS may represent an early marker of immune system dysregulation in young, otherwise healthy, adults with MDD.NEW & NOTEWORTHY To our knowledge, we provide the first evidence of augmented T-cell mitochondrial reactive oxygen species (T-cell mitoROS) in young, otherwise healthy adults with MDD. Although the elevated T-cell mitoROS did not correspond to a proinflammatory profile, these findings suggest that elevated T-cell mitoROS may be an early marker of immune system dysregulation in young adults with MDD.


Subject(s)
Depressive Disorder, Major/immunology , Mitochondria/chemistry , Reactive Oxygen Species/analysis , T-Lymphocytes/ultrastructure , Adult , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , COVID-19/immunology , COVID-19/psychology , Cytokines , Female , Humans , Ki-1 Antigen/analysis , Male , SARS-CoV-2 , Severity of Illness Index , Young Adult
9.
J Physiol ; 599(16): 3973-3991, 2021 08.
Article in English | MEDLINE | ID: mdl-34164826

ABSTRACT

KEY POINTS: Increased large artery stiffness and impaired endothelium-dependent dilatation occur with advanced age. We sought to determine whether T cells mechanistically contribute to age-related arterial dysfunction. We found that old mice exhibited greater proinflammatory T cell accumulation around both the aorta and mesenteric arteries. Pharmacologic depletion or genetic deletion of T cells in old mice resulted in ameliorated large artery stiffness and greater endothelium-dependent dilatation compared with mice with T cells intact. ABSTRACT: Ageing of the arteries is characterized by increased large artery stiffness and impaired endothelium-dependent dilatation. T cells contribute to hypertension in acute rodent models but whether they contribute to chronic age-related arterial dysfunction is unknown. To determine whether T cells directly mediate age-related arterial dysfunction, we examined large elastic artery and resistance artery function in young (4-6 months) and old (22-24 months) wild-type mice treated with anti-CD3 F(ab'2) fragments to deplete T cells (150 µg, i.p. every 7 days for 28 days) or isotype control fragments. Old mice exhibited greater numbers of T cells in both aorta and mesenteric vasculature when compared with young mice. Old mice treated with anti-CD3 fragments exhibited depletion of T cells in blood, spleen, aorta and mesenteric vasculature. Old mice also exhibited greater numbers of aortic and mesenteric IFN-γ and TNF-α-producing T cells when compared with young mice. Old control mice exhibited greater large artery stiffness and impaired resistance artery endothelium-dependent dilatation in comparison with young mice. In old mice, large artery stiffness was ameliorated with anti-CD3 treatment. Anti-CD3-treated old mice also exhibited greater endothelium-dependent dilatation than age-matched controls. We also examined arterial function in young and old Rag-1-/- mice, which lack lymphocytes. Rag-1-/- mice exhibited blunted increases in large artery stiffness with age compared with wild-type mice. Old Rag-1-/- mice also exhibited greater endothelium-dependent dilatation compared with old wild-type mice. Collectively, these results demonstrate that T cells play an important role in age-related arterial dysfunction.


Subject(s)
Vascular Stiffness , Aging , Animals , Endothelium, Vascular , Mesenteric Arteries , Mice , T-Lymphocytes , Vasodilation
10.
Geroscience ; 43(3): 1331-1347, 2021 06.
Article in English | MEDLINE | ID: mdl-33893902

ABSTRACT

Both glucose tolerance and adaptive immune function exhibit significant age-related alterations. The influence of the immune system on obesity-associated glucose intolerance is well characterized; however, whether the immune system contributes to age-related glucose intolerance is not as well understood. Here, we report that advancing age results in an increase in T cell infiltration in the epididymal white adipose tissue (eWAT), liver, and skeletal muscle. Subtype analyses show that both CD4+, CD8+ T cells are greater with advancing age in each of these tissues and that aging results in a blunted CD4 to CD8 ratio. Anti-CD3 F(ab')2 fragments depleted CD4+ and CD8+ cells in eWAT, CD4+ cells only in the liver, and did not deplete quadriceps T cells. In old mice, T cells producing both interferon-γ and tumor necrosis factor-α are accumulated in the eWAT and liver, and a greater proportion of skeletal muscle T cells produced interferon-γ. Aging resulted in increased proportion and numbers of T regulatory cells in eWAT, but not in the liver or muscle. Aging also resulted in greater numbers of eWAT and quadriceps CD206- macrophages and eWAT, liver and quadriceps B cells; neither cell type was altered by anti-CD3 treatment. Anti-CD3 treatment improved glucose tolerance in old mice and was accompanied by improved signaling related to liver and skeletal muscle insulin utilization and decreased gluconeogenesis-related gene expression in the liver. Our findings indicate a critical role of the adaptive immune system in the age-related metabolic dysfunction.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Adipose Tissue, White , Animals , Lymphocyte Depletion , Mice , Mice, Inbred C57BL
11.
Exp Physiol ; 104(10): 1455-1471, 2019 10.
Article in English | MEDLINE | ID: mdl-31325339

ABSTRACT

NEW FINDINGS: What is the topic of this review? This review summarizes and synthesizes what is known about the contribution of inflammation to age-related arterial dysfunction. What advances does it highlight? This review details observational evidence for the relationship of age-related inflammation and arterial dysfunction, insight from autoimmune inflammatory diseases and their effects on arterial function, interventional evidence linking inflammation and age-related arterial dysfunction, insight into age-related arterial inflammation from preclinical models and interventions to ameliorate age-related inflammation and arterial dysfunction. ABSTRACT: Advanced age is a primary risk factor for cardiovascular disease, the leading cause of death in the industrialized world. Two major components of arterial ageing are stiffening of the large arteries and impaired endothelium-dependent dilatation in multiple vascular beds. These two alterations are major contributors to the development of overt cardiovascular disease. Increasing inflammation with advanced age is likely to play a role in this arterial dysfunction. The purpose of this review is to synthesize what is known about inflammation and its relationship to age-related arterial dysfunction. This review discusses both the initial observational evidence for the relationship of age-related inflammation and arterial dysfunction and the evidence that inflammatory autoimmune diseases are associated with a premature arterial ageing phenotype. We next discuss interventional and mechanistic evidence linking inflammation and age-related arterial dysfunction in older adults. We also attempt to summarize the relevant evidence from preclinical models. Lastly, we discuss interventions in both humans and animals that have been shown to ameliorate age-related arterial inflammation and dysfunction. The available evidence provides a strong basis for the role of inflammation in both large artery stiffening and impairment of endothelium-dependent dilatation; however, the specific inflammatory mediators, the initiating factors and the relative importance of the endothelium, smooth muscle cells, perivascular adipose tissue and immune cells in arterial inflammation are not well understood. With the expansion of the ageing population, ameliorating age-related arterial inflammation represents an important potential strategy for preserving vascular health in the elderly.


Subject(s)
Aging/pathology , Arteries/pathology , Inflammation/pathology , Animals , Cardiovascular Diseases , Endothelium, Vascular/pathology , Humans
12.
J Mol Cell Cardiol ; 127: 74-82, 2019 02.
Article in English | MEDLINE | ID: mdl-30502348

ABSTRACT

Age-related vascular dysfunction in large elastic and resistance arteries is associated with reductions in microvascular perfusion and elevations in blood pressure. Recent evidence indicates that telomere uncapping-induced senescence in vascular cells may be an important source of oxidative stress and vascular dysfunction in aging, but the causal relationship between these processes has yet to be elucidated. To test this important unexplored hypothesis, we measured arterial senescence signaling and oxidative stress, carotid and mesenteric artery endothelium-dependent vasodilatory capacity, markers of mesenteric microvascular perfusion and endothelial glycocalyx deterioration, and blood pressure in a novel mouse model of Cre-inducible whole body Trf2 deletion and telomere uncapping. Trf2 deletion led to a 320% increase in arterial senescence signaling (P < .05). There was a concurrent 29% and 22% reduction in peak endothelium-dependent vasodilation in carotid and mesenteric arteries, respectively, as well as a 63% reduction in mesenteric microvascular endothelial glycocalyx thickness (all P ≤ .01). Mesenteric microvascular perfusion was reduced by 8% and systolic blood pressure was increased by 9% following Trf2 deletion (both P < .05). Trf2 deletion also led to a pro-oxidative arterial phenotype characterized by increased in NADPH oxidase gene expression; a 210% increase in superoxide levels that was partly dependent on NADPH oxidase activity; and an oxidative stress mediated reduction in carotid artery vasodilation (all P ≤ .05). Collectively, our findings demonstrate that induced Trf2 deletion leads to telomere uncapping, increased senescence signaling, and oxidative stress mediated functional impairments in the vasculature similar to those seen in human aging.


Subject(s)
Aging/metabolism , Arteries/metabolism , Cellular Senescence , Gene Deletion , Oxidative Stress , Signal Transduction , Telomere/metabolism , Telomeric Repeat Binding Protein 2/deficiency , Adipose Tissue/metabolism , Animals , Blood Pressure , Body Weight , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Glycocalyx/metabolism , Mice , Microvessels/metabolism , Perfusion , Phenotype , Telomere Homeostasis , Telomeric Repeat Binding Protein 2/metabolism , Vasodilation
13.
Exp Gerontol ; 109: 99-107, 2018 08.
Article in English | MEDLINE | ID: mdl-28012941

ABSTRACT

Age-related arterial inflammation is associated with dysfunction of the arteries and increased risk for cardiovascular disease. To determine if aging increases arterial immune cell infiltration as well as the populations of immune cells principally involved, we tested the hypothesis that large elastic and resistance arteries in old mice would exhibit increased immune cell infiltration compared to young controls. Additionally, we hypothesized that vasoprotective lifestyle interventions such as lifelong caloric restriction or 8weeks of voluntary wheel running would attenuate age-related arterial immune cell infiltration. The aorta and mesenteric vasculature with surrounding perivascular adipose was excised from young normal chow (YNC, 4-6months, n=10), old normal chow (ONC, 28-29months, n=11), old caloric restricted (OCR, 28-29months, n=9), and old voluntary running (OVR, 28-29months, n=5) mice and digested to a single cell suspension. The cells were then labeled with antibodies against CD45 (total leukocytes), CD3 (pan T cells), CD4 (T helper cells), CD8 (cytotoxic T cells), CD19 (B cells), CD11b, and F4/80 (macrophages) and analyzed by flow cytometry. Total leukocytes, T cells (both CD4+ and CD8+ subsets), B cells, and macrophages in both aorta and mesentery were all 5- to 6-fold greater in ONC compared to YNC. Age-related increases in T cell (both CD4+ and CD8+), B cell, and macrophage infiltration in aorta were abolished in OCR mice. OVR mice exhibited 50% lower aortic T cell and normalized macrophage infiltration. B cell infiltration was not affected by VR. Age-related mesenteric CD8+ T cell and macrophage infiltration was normalized in OCR and OVR mice compared to young mice, whereas B cell infiltration was normalized by CR but not VR. Splenic CD4+ T cells from ONC mice exhibited a 3-fold increase in gene expression for the T helper (Th) 1 transcription factor, Tbet, and a 4-fold increase in FoxP3, a T regulatory cell transcription factor, compared to YNC. Splenic B cells and mesenteric macrophages from old mice exhibited decreased proinflammatory cytokine gene expression regardless of treatment group. These results demonstrate that aging is associated with infiltration of immune cells around both the large-elastic and resistance arteries and that the vasoprotective lifestyle interventions, CR and VR, can ameliorate age-related arterial immune cell infiltration.


Subject(s)
Aging/immunology , Arteries/immunology , Caloric Restriction , Physical Conditioning, Animal , Animals , Leukocytes/physiology , Macrophages/physiology , Male , Mice , Vascular Diseases/etiology
14.
Cytokine ; 96: 102-106, 2017 08.
Article in English | MEDLINE | ID: mdl-28390264

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death in the industrialized world. Aging is the most predictive risk factor for CVD and is associated with arterial inflammation which contributes to increased CVD risk. Although age-related arterial inflammation has been described in both humans and animals, only a limited number of inflammatory mediators, cytokines and chemokines have been identified. In this investigation we sought to determine whether lifespan extending interventions, including crowded litter early life nutrient deprivation (CL), traditional lifelong caloric restriction (CR) and lifelong Rapamycin treatment (Rap) would attenuate age-related arterial inflammation using multi analyte profiling. Aortas from Young (4-6months), Old (22months), Old CL, Old CR and Old Rap mice were homogenized and cytokine concentrations were assessed using Luminex Multi Analyte Profiling. Chemokines involved in immune cell recruitment, such as CCL2, CXCL9, CXCL10, GMCSF and MCSF, were increased in Old vs. Young (p<0.05). The age-related increase of CXCL10 was prevented by CR (p<0.05 vs. Old). MSCF concentrations were lower in aortas of Rap treated mice (p<0.05 vs. Old). Interleukins (IL), IL-1α, IL-1ß and IL-10, were also greater in Old vs. Young mice (p<0.05). These data demonstrate selected lifespan extending interventions can prevent or limit age-related increases in selected aortic chemokines.


Subject(s)
Aging , Arteries/immunology , Chemokine CXCL10/blood , Macrophage Colony-Stimulating Factor/blood , Animals , Caloric Restriction , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Chemokine CCL2/biosynthesis , Chemokine CCL4/biosynthesis , Chemokine CXCL10/immunology , Chemokines/biosynthesis , Chemokines/immunology , Cytokines/biosynthesis , Cytokines/immunology , Early Medical Intervention , Interleukin-10/biosynthesis , Interleukin-1beta/blood , Interleukins/biosynthesis , Interleukins/immunology , Macrophage Colony-Stimulating Factor/immunology , Male , Mice , Sirolimus/therapeutic use
15.
Aging Cell ; 16(1): 17-26, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27660040

ABSTRACT

Inhibition of mammalian target of rapamycin, mTOR, extends lifespan and reduces age-related disease. It is not known what role mTOR plays in the arterial aging phenotype or if mTOR inhibition by dietary rapamycin ameliorates age-related arterial dysfunction. To explore this, young (3.8 ± 0.6 months) and old (30.3 ± 0.2 months) male B6D2F1 mice were fed a rapamycin supplemented or control diet for 6-8 weeks. Although there were few other notable changes in animal characteristics after rapamycin treatment, we found that glucose tolerance improved in old mice, but was impaired in young mice, after rapamycin supplementation (both P < 0.05). Aging increased mTOR activation in arteries evidenced by elevated S6K phosphorylation (P < 0.01), and this was reversed after rapamycin treatment in old mice (P < 0.05). Aging was also associated with impaired endothelium-dependent dilation (EDD) in the carotid artery (P < 0.05). Rapamycin improved EDD in old mice (P < 0.05). Superoxide production and NADPH oxidase expression were higher in arteries from old compared to young mice (P < 0.05), and rapamycin normalized these (P < 0.05) to levels not different from young mice. Scavenging superoxide improved carotid artery EDD in untreated (P < 0.05), but not rapamycin-treated, old mice. While aging increased large artery stiffness evidenced by increased aortic pulse-wave velocity (PWV) (P < 0.01), rapamycin treatment reduced aortic PWV (P < 0.05) and collagen content (P < 0.05) in old mice. Aortic adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and expression of the cell cycle-related proteins PTEN and p27kip were increased with rapamycin treatment in old mice (all P < 0.05). Lastly, aging resulted in augmentation of the arterial senescence marker, p19 (P < 0.05), and this was ameliorated by rapamycin treatment (P < 0.05). These results demonstrate beneficial effects of rapamycin treatment on arterial function in old mice and suggest these improvements are associated with reduced oxidative stress, AMPK activation and increased expression of proteins involved in the control of the cell cycle.


Subject(s)
Aging/pathology , Cell Cycle/drug effects , Cellular Senescence/drug effects , Dietary Supplements , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Oxidative Stress/drug effects , Sirolimus/pharmacology , Adenylate Kinase/metabolism , Animals , Arteries/drug effects , Arteries/pathology , Arteries/physiopathology , Biomarkers/metabolism , Blood Glucose/metabolism , Body Weight/drug effects , Cell Cycle Proteins/metabolism , Endothelium, Vascular/drug effects , Homeostasis/drug effects , Insulin/blood , Insulin Resistance , Male , Mice, Inbred C57BL , Organ Size/drug effects , TOR Serine-Threonine Kinases/metabolism , Vascular Stiffness/drug effects , Vasodilation/drug effects
16.
Hypertension ; 64(5): 1108-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25259750

ABSTRACT

Recent studies have emphasized a role of adaptive immunity, and particularly T cells, in the genesis of hypertension. We sought to determine the T-cell subtypes that contribute to hypertension and renal inflammation in angiotensin II-induced hypertension. Using T-cell receptor spectratyping to examine T-cell receptor usage, we demonstrated that CD8(+) cells, but not CD4(+) cells, in the kidney exhibited altered T-cell receptor transcript lengths in Vß3, 8.1, and 17 families in response to angiotensin II-induced hypertension. Clonality was not observed in other organs. The hypertension caused by angiotensin II in CD4(-/-) and MHCII(-/-) mice was similar to that observed in wild-type mice, whereas CD8(-/-) mice and OT1xRAG-1(-/-) mice, which have only 1 T-cell receptor, exhibited a blunted hypertensive response to angiotensin II. Adoptive transfer of pan T cells and CD8(+) T cells but not CD4(+)/CD25(-) cells conferred hypertension to RAG-1(-/-) mice. In contrast, transfer of CD4(+)/CD25(+) cells to wild-type mice receiving angiotensin II decreased blood pressure. Mice treated with angiotensin II exhibited increased numbers of kidney CD4(+) and CD8(+) T cells. In response to a sodium/volume challenge, wild-type and CD4(-/-) mice infused with angiotensin II retained water and sodium, whereas CD8(-/-) mice did not. CD8(-/-) mice were also protected against angiotensin-induced endothelial dysfunction and vascular remodeling in the kidney. These data suggest that in the development of hypertension, an oligoclonal population of CD8(+) cells accumulates in the kidney and likely contributes to hypertension by contributing to sodium and volume retention and vascular rarefaction.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/physiology , Hypertension/pathology , Hypertension/physiopathology , Kidney/immunology , Kidney/pathology , Oligoclonal Bands/physiology , Adaptive Immunity/physiology , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , CD4 Antigens/genetics , CD4 Antigens/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/physiology , CD8 Antigens/genetics , CD8 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Hypertension/chemically induced , Kidney/blood supply , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/physiology , Male , Mice , Mice, Knockout , Vascular Remodeling/drug effects
17.
J Clin Invest ; 124(10): 4642-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25244096

ABSTRACT

Oxidative damage and inflammation are both implicated in the genesis of hypertension; however, the mechanisms by which these stimuli promote hypertension are not fully understood. Here, we have described a pathway in which hypertensive stimuli promote dendritic cell (DC) activation of T cells, ultimately leading to hypertension. Using multiple murine models of hypertension, we determined that proteins oxidatively modified by highly reactive γ-ketoaldehydes (isoketals) are formed in hypertension and accumulate in DCs. Isoketal accumulation was associated with DC production of IL-6, IL-1ß, and IL-23 and an increase in costimulatory proteins CD80 and CD86. These activated DCs promoted T cell, particularly CD8+ T cell, proliferation; production of IFN-γ and IL-17A; and hypertension. Moreover, isoketal scavengers prevented these hypertension-associated events. Plasma F2-isoprostanes, which are formed in concert with isoketals, were found to be elevated in humans with treated hypertension and were markedly elevated in patients with resistant hypertension. Isoketal-modified proteins were also markedly elevated in circulating monocytes and DCs from humans with hypertension. Our data reveal that hypertension activates DCs, in large part by promoting the formation of isoketals, and suggest that reducing isoketals has potential as a treatment strategy for this disease.


Subject(s)
Dendritic Cells/immunology , Hypertension/pathology , Lymphocyte Activation , T-Lymphocytes/cytology , Aged , Aldehydes/chemistry , Angiotensin II/metabolism , Animals , Antigen-Presenting Cells/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Cell Proliferation , Cohort Studies , Dendritic Cells/cytology , Female , Gene Expression Regulation , Humans , Inflammation , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Interleukin-23/metabolism , Interleukin-6/metabolism , Kidney/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Oxidative Stress , Oxygen/metabolism , Superoxides/metabolism
18.
Adv Physiol Educ ; 38(1): 20-4, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24585465

ABSTRACT

While hypertension has predominantly been attributed to perturbations of the vasculature, kidney, and central nervous system, research for almost 50 yr has shown that the immune system also contributes to this disease. Inflammatory cells accumulate in the kidneys and vasculature of humans and experimental animals with hypertension and likely contribute to end-organ damage. We and others have shown that mice lacking adaptive immune cells, including recombinase-activating gene-deficient mice and rats and mice with severe combined immunodeficiency have blunted hypertension to stimuli such as ANG II, high salt, and norepinephrine. Adoptive transfer of T cells restores the blood pressure response to these stimuli. Agonistic antibodies to the ANG II receptor, produced by B cells, contribute to hypertension in experimental models of preeclampsia. The central nervous system seems important in immune cell activation, because lesions in the anteroventral third ventricle block hypertension and T cell activation in response to ANG II. Likewise, genetic manipulation of reactive oxygen species in the subfornical organ modulates both hypertension and immune cell activation. Current evidence indicates that the production of cytokines, including tumor necrosis factor-α, interleukin-17, and interleukin-6, contribute to hypertension, likely via effects on both the kidney and vasculature. In addition, the innate immune system also appears to contribute to hypertension. We propose a working hypothesis linking the sympathetic nervous system, immune cells, production of cytokines, and, ultimately, vascular and renal dysfunction, leading to the augmentation of hypertension. Studies of immune cell activation will clearly be useful in understanding this common yet complex disease.


Subject(s)
Blood Pressure , Hypertension/immunology , Immune System/immunology , Animals , B-Lymphocytes/immunology , Central Nervous System/immunology , Central Nervous System/physiopathology , Cytokines/metabolism , Humans , Hypertension/metabolism , Hypertension/physiopathology , Immune System/metabolism , Immune System/physiopathology , Immunity, Innate , Inflammation Mediators/metabolism , Signal Transduction , Sympathetic Nervous System/immunology , Sympathetic Nervous System/physiopathology , T-Lymphocytes/immunology
19.
Circ Res ; 114(4): 616-25, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24347665

ABSTRACT

RATIONALE: Aortic stiffening commonly occurs in hypertension and further elevates systolic pressure. Hypertension is also associated with vascular inflammation and increased mechanical stretch. The interplay between inflammation, mechanical stretch, and aortic stiffening in hypertension remains undefined. OBJECTIVE: Our aim was to determine the role of inflammation and mechanical stretch in aortic stiffening. METHODS AND RESULTS: Chronic angiotensin II infusion caused marked aortic adventitial collagen deposition, as quantified by Masson trichrome blue staining and biochemically by hydroxyproline content, in wild-type but not in recombination activating gene-1-deficient mice. Aortic compliance, defined by ex vivo measurements of stress-strain curves, was reduced by chronic angiotensin II infusion in wild-type mice (P<0.01) but not in recombination activating gene-1-deficient mice (P<0.05). Adoptive transfer of T-cells to recombination activating gene-1-deficient mice restored aortic collagen deposition and stiffness to values observed in wild-type mice. Mice lacking the T-cell-derived cytokine interleukin 17a were also protected against aortic stiffening. In additional studies, we found that blood pressure normalization by treatment with hydralazine and hydrochlorothiazide prevented angiotensin II-induced vascular T-cell infiltration, aortic stiffening, and collagen deposition. Finally, we found that mechanical stretch induces the expression of collagen 1α1, 3α1, and 5a1 in cultured aortic fibroblasts in a p38 mitogen-activated protein kinase-dependent fashion, and that inhibition of p38 prevented angiotensin II-induced aortic stiffening in vivo. Interleukin 17a also induced collagen 3a1 expression via the activation of p38 mitogen-activated protein kinase. CONCLUSIONS: Our data define a pathway in which inflammation and mechanical stretch lead to vascular inflammation that promotes collagen deposition. The resultant increase in aortic stiffness likely further worsens systolic hypertension and its attendant end-organ damage.


Subject(s)
Aortic Diseases/metabolism , Hypertension/metabolism , Inflammation/metabolism , Vascular Stiffness/physiology , Vasculitis/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adoptive Transfer , Angiotensin II/pharmacology , Animals , Aortic Diseases/physiopathology , CD4 Antigens/genetics , CD8 Antigens/genetics , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Elastin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Hypertension/chemically induced , Hypertension/physiopathology , Interleukin-17/genetics , Male , Mice , Mice, Knockout , Stress, Mechanical , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Vasculitis/physiopathology , Vasoconstrictor Agents/pharmacology
20.
Eur J Appl Physiol ; 113(8): 2039-46, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23563601

ABSTRACT

We tested the hypothesis that impaired nitric oxide (NO)-mediated, endothelium-dependent dilation in aged soleus muscle feed arteries (SFA) is due to an age-related decline in the potential for PI3-kinase (PI3K)/protein kinase B (Akt)-dependent phosphorylation of endothelial NO synthase (eNOS) on serine residue 1177 (p-eNOS(ser1177)). SFA from young (4 months) and old (24 months) Fischer 344 rats were cannulated for examination of endothelium-dependent [flow or acetylcholine (ACh)] and endothelium-independent [sodium nitroprusside (SNP)] vasodilator function. To determine the mechanism by which aging affected vasodilation to flow and ACh, vasodilator responses were assessed in the presence of N (ω)-nitro-L-arginine (L-NNA, to inhibit NOS), LY-294002 (to inhibit PI3K), or 1L6-hydroxymethyl-chiro-inositol-2-(R)-2-O-methyl-3-O-octadecyl-sn-glycerocarbonate (AktI, to inhibit Akt). Flow- and ACh-induced vasodilator responses were significantly impaired in old SFA, whereas endothelium-independent dilation to SNP was not compromised. Age-group differences in flow- and ACh-induced dilations were abolished in the presence of L-NNA, LY-294002, or AktI. In a separate experiment, SFA were cannulated and stimulated with ACh (10(-4) M, 3 min), flow (60 µl/min, 5 min), or remained unstimulated (3 min). SFA were removed from the pipettes and immunoblot analysis was used to assess ACh- and flow-stimulated phosphorylation of eNOS on ser(1177). Stimulation with ACh or flow increased phosphorylation of eNOS on ser(1177) in young (not old) SFA. Preincubation of young SFA with LY-294002, abolished the ACh-induced phosphorylation of eNOS in young SFA. Collectively, these results indicate that impaired NO-mediated, endothelium-dependent dilation in old SFA is due, in part, to an impaired potential for PI3K/Akt-dependent phosphorylation of eNOS on ser(1177).


Subject(s)
Aging , Endothelium, Vascular/physiology , Muscle, Skeletal/blood supply , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vasodilation , Animals , Arteries/anatomy & histology , Arteries/physiology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Hindlimb/blood supply , Male , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats , Rats, Inbred F344 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...