Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 324(2): R161-R170, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36534590

ABSTRACT

Bsardet Biedl syndrome (BBS) is a genetic condition associated with various clinical features including cutaneous disorders and certain autoimmune and inflammatory diseases pointing to a potential role of BBS proteins in the regulation of immune function. BBS1 protein, which is a key component of the BBSome, a protein complex involved in the regulation of cilia function and other cellular processes, has been implicated in the immune synapse assembly by promoting the centrosome polarization to the antigen-presenting cells. Here, we assessed the effect of disrupting the BBSome, through Bbs1 gene deletion, in T cells. Interestingly, mice lacking the Bbs1 gene specifically in T cells (T-BBS1-/-) displayed normal body weight, adiposity, and glucose handling, but have smaller spleens. However, T-BBS1-/- mice had no change in the proportion and absolute number of B cells and T cells in the spleen and lymph nodes. There was also no alteration in the CD4/CD8 lineage commitment or survival in the thymus of T-BBS1-/- mice. On the other hand, T-BBS1-/- mice treated with Imiquimod dermally exhibited a significantly higher percentage of CD3-positive splenocytes that was due to CD4 but not CD8 T cell predominance. Notably, we found that T-BBS1-/- mice had significantly decreased wound closure, an effect that was more pronounced in males indicating that the BBSome plays an important role in T cell-mediated skin repair. Together, these findings implicate the BBSome in the regulation of selective functions of T cells.


Subject(s)
Cilia , Microtubule-Associated Proteins , Animals , Male , Mice , Adiposity , Cilia/metabolism , Cilia/pathology , Immunity/genetics , Microtubule-Associated Proteins/genetics , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism
2.
Pediatr Dermatol ; 39(1): 145-146, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34888940

ABSTRACT

Actinic prurigo is a rare pruritic photodermatosis. We report the use of Polypodium leucotomos extract in an 11-year-old female patient with actinic prurigo, resulting in a significant attenuation of her disease without development of adverse effects to date. Polypodium leucotomos exerts a pleiotropic immunomodulatory and antioxidant effect by shifting the balance from pro- to an antiinflammatory cytokine environment. This counteracts the effects of UV-induced cellular damage characteristic of photodermatoses.


Subject(s)
Polypodium , Prurigo , Antioxidants , Child , Dietary Supplements , Female , Humans , Photosensitivity Disorders , Plant Extracts/therapeutic use , Prurigo/drug therapy , Skin Diseases, Genetic
3.
Physiol Genomics ; 48(7): 491-501, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27199455

ABSTRACT

Peroxisome proliferator-activated receptor-γ (PPARγ), a master regulator of adipogenesis, was recently shown to affect energy homeostasis through its actions in the brain. Deletion of PPARγ in mouse brain, and specifically in the pro-opiomelanocortin (POMC) neurons, results in resistance to diet-induced obesity. To study the mechanisms by which PPARγ in POMC neurons controls energy balance, we constructed a Cre-recombinase-dependent conditionally activatable transgene expressing either wild-type (WT) or dominant-negative (P467L) PPARγ and the tdTomato reporter. Inducible expression of both forms of PPARγ was validated in cells in culture, in liver of mice infected with an adenovirus expressing Cre-recombinase (AdCre), and in the brain of mice expressing Cre-recombinase either in all neurons (NES(Cre)/PPARγ-P467L) or selectively in POMC neurons (POMC(Cre)/PPARγ-P467L). Whereas POMC(Cre)/PPARγ-P467L mice exhibited a normal pattern of weight gain when fed 60% high-fat diet, they exhibited increased weight gain and fat mass accumulation in response to a 10% fat isocaloric-matched control diet. POMC(Cre)/PPARγ-P467L mice were leptin sensitive on control diet but became leptin resistant when fed 60% high-fat diet. There was no difference in body weight between POMC(Cre)/PPARγ-WT mice and controls in response to 60% high-fat diet. However, POMC(Cre)/PPARγ-WT, but not POMC(Cre)/PPARγ-P467L, mice increased body weight in response to rosiglitazone, a PPARγ agonist. These observations support the concept that alterations in PPARγ-driven mechanisms in POMC neurons can play a role in the regulation of metabolic homeostasis under certain dietary conditions.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Neurons/metabolism , PPAR gamma/metabolism , Pro-Opiomelanocortin/metabolism , 3T3 Cells , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , Body Weight/drug effects , Body Weight/physiology , Brain/drug effects , Brain/metabolism , Brain/physiology , Cell Line , Diet, High-Fat/methods , Energy Metabolism/drug effects , Female , HEK293 Cells , Homeostasis/drug effects , Humans , Leptin/metabolism , Male , Mice , Neurons/drug effects , Obesity/metabolism , Obesity/physiopathology , PPAR gamma/agonists , Rosiglitazone , Thiazolidinediones/pharmacology , Weight Gain/drug effects , Weight Gain/physiology
4.
Am J Physiol Heart Circ Physiol ; 310(1): H39-48, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26566726

ABSTRACT

Loss of peroxisome proliferator-activated receptor (PPAR)-γ function in the vascular endothelium enhances atherosclerosis and NF-κB target gene expression in high-fat diet-fed apolipoprotein E-deficient mice. The mechanisms by which endothelial PPAR-γ regulates inflammatory responses and protects against atherosclerosis remain unclear. To assess functional interactions between PPAR-γ and inflammation, we used a model of IL-1ß-induced aortic dysfunction in transgenic mice with endothelium-specific overexpression of either wild-type (E-WT) or dominant negative PPAR-γ (E-V290M). IL-1ß dose dependently decreased IκB-α, increased phospho-p65, and increased luciferase activity in the aorta of NF-κB-LUC transgenic mice. IL-1ß also dose dependently reduced endothelial-dependent relaxation by ACh. The loss of ACh responsiveness was partially improved by pretreatment of the vessels with the PPAR-γ agonist rosiglitazone or in E-WT. Conversely, IL-1ß-induced endothelial dysfunction was worsened in the aorta from E-V290M mice. Although IL-1ß increased the expression of NF-κB target genes, NF-κB p65 inhibitor did not alleviate endothelial dysfunction induced by IL-1ß. Tempol, a SOD mimetic, partially restored ACh responsiveness in the IL-1ß-treated aorta. Notably, tempol only modestly improved protection in the E-WT aorta but had an increased protective effect in the E-V290M aorta compared with the aorta from nontransgenic mice, suggesting that PPAR-γ-mediated protection involves antioxidant effects. IL-1ß increased ROS and decreased the phospho-endothelial nitric oxide synthase (Ser(1177))-to-endothelial nitric oxide synthase ratio in the nontransgenic aorta. These effects were completely abolished in the aorta with endothelial overexpression of WT PPAR-γ but were worsened in the aorta with E-V290M even in the absence of IL-1ß. We conclude that PPAR-γ protects against IL-1ß-mediated endothelial dysfunction through a reduction of oxidative stress responses but not by blunting IL-1ß-mediated NF-κB activity.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Interleukin-1beta/pharmacology , Oxidative Stress/drug effects , PPAR gamma/metabolism , Animals , Antioxidants/pharmacology , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Gene Expression Regulation , Genotype , Humans , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , NF-KappaB Inhibitor alpha , Nitric Oxide Synthase Type III/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , Phenotype , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Curr Hypertens Rep ; 17(12): 89, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26462805

ABSTRACT

Dysregulation of peroxisome proliferator-activated receptor gamma (PPARγ) activity leads to significant alterations in cardiovascular and metabolic regulation. This is most keenly observed by the metabolic syndrome-like phenotypes exhibited by patients carrying mutations in PPARγ. We will summarize recent findings regarding mechanisms of PPARγ regulation in the cardiovascular and nervous systems focusing largely on PPARγ in the smooth muscle, endothelium, and brain. Canonically, PPARγ exerts its effects by regulating the expression of target genes in these cells, and we will discuss mechanisms by which PPARγ targets in the vasculature regulate cardiovascular function. We will also discuss emerging evidence that PPARγ in the brain is a mediator of appetite and obesity. Finally, we will briefly review how novel PPARγ activators control posttranslational modifications of PPARγ and their prospects to offer new therapeutic options for treatment of metabolic diseases without the adverse side effects of thiazolidinediones which strongly activate transcriptional activity of PPARγ.


Subject(s)
Hypertension/metabolism , Metabolic Syndrome/metabolism , PPAR gamma/metabolism , Animals , Blood Pressure , Central Nervous System/metabolism , Humans , Hypertension/drug therapy , Hypertension/etiology , Metabolic Syndrome/complications , Metabolic Syndrome/drug therapy , Obesity/metabolism
6.
JAMA Dermatol ; 160(8): 799-800, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38985483

ABSTRACT

This Viewpoint compares 2 clinical trials involving immune checkpoint inhibitors in kidney transplant recipients.


Subject(s)
Kidney Transplantation , Skin Neoplasms , Humans , Skin Neoplasms/pathology , Immunotherapy/methods , Transplant Recipients/statistics & numerical data
7.
Hypertension ; 70(1): 174-182, 2017 07.
Article in English | MEDLINE | ID: mdl-28507170

ABSTRACT

Selective expression of dominant negative (DN) peroxisome proliferator-activated receptor γ (PPARγ) in vascular smooth muscle cells (SMC) results in hypertension, atherosclerosis, and increased nuclear factor-κB (NF-κB) target gene expression. Mesenteric SMC were cultured from mice designed to conditionally express wild-type (WT) or DN-PPARγ in response to Cre recombinase to determine how SMC PPARγ regulates expression of NF-κB target inflammatory genes. SMC-specific overexpression of WT-PPARγ or agonist-induced activation of endogenous PPARγ blunted tumor necrosis factor α (TNF-α)-induced NF-κB target gene expression and activity of an NF-κB-responsive promoter. TNF-α-induced gene expression responses were enhanced by DN-PPARγ in SMC. Although expression of NF-κB p65 was unchanged, nuclear export of p65 was accelerated by WT-PPARγ and prevented by DN-PPARγ in SMC. Leptomycin B, a nuclear export inhibitor, blocked p65 nuclear export and inhibited the anti-inflammatory action of PPARγ. Consistent with a role in facilitating p65 nuclear export, WT-PPARγ coimmunoprecipitated with p65, and WT-PPARγ was also exported from the nucleus after TNF-α treatment. Conversely, DN-PPARγ does not bind to p65 and was retained in the nucleus after TNF-α treatment. Transgenic mice expressing WT-PPARγ or DN-PPARγ specifically in SMC (S-WT or S-DN) were bred with mice expressing luciferase controlled by an NF-κB-responsive promoter to assess effects on NF-κB activity in whole tissue. TNF-α-induced NF-κB activity was decreased in aorta and carotid artery from S-WT but was increased in vessels from S-DN mice. We conclude that SMC PPARγ blunts expression of proinflammatory genes by inhibition of NF-κB activity through a mechanism promoting nuclear export of p65, which is abolished by DN mutation in PPARγ.


Subject(s)
Hypertension , Muscle, Smooth, Vascular , NF-kappa B , PPAR gamma/genetics , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Fatty Acids, Unsaturated/pharmacology , Hypertension/genetics , Hypertension/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Mutation , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Endocrinology ; 157(11): 4266-4275, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27575030

ABSTRACT

Peroxisome proliferator activated receptor (PPARγ) is a nuclear receptor transcription factor that regulates adipogenesis and energy homeostasis. Recent studies suggest PPARγ may mediate some of its metabolic effects through actions in the brain. We used a Cre-recombinase-dependent (using NestinCre) conditionally activatable transgene expressing either wild-type (WT) or dominant-negative (P467L) PPARγ to examine mechanisms by which PPARγ in the nervous system controls energy balance. Inducible expression of PPARγ was evident throughout the brain. Expression of 2 PPARγ target genes, aP2 and CD36, was induced by WT but not P467L PPARγ in the brain. Surprisingly, NesCre/PPARγ-WT mice exhibited severe microcephaly and brain malformation, suggesting that PPARγ can modulate brain development. On the contrary, NesCre/PPARγ-P467L mice exhibited blunted weight gain to high-fat diet, which correlated with a decrease in lean mass and tissue masses, accompanied by elevated plasma GH, and depressed plasma IGF-1, indicative of GH resistance. There was no expression of the transgene in the pancreas but both fasting plasma glucose, and fed and fasted plasma insulin levels were markedly decreased. NesCre/PPARγ-P467L mice fed either control diet or high-fat diet displayed impaired glucose tolerance yet exhibited increased sensitivity to exogenous insulin and increased insulin receptor signaling in white adipose tissue, liver, and skeletal muscle. These observations support the concept that alterations in PPARγ-driven mechanisms in the nervous system play a role in the regulation of growth and glucose metabolic homeostasis.


Subject(s)
Brain/growth & development , Brain/metabolism , PPAR gamma/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Body Composition/genetics , Body Composition/physiology , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Energy Metabolism/physiology , Fasting/blood , Glucose/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Resistance/genetics , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Transgenic , Mutation , PPAR gamma/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
9.
Hypertension ; 67(1): 214-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26597823

ABSTRACT

Loss of peroxisome proliferator-activated receptor-γ (PPARγ) function causes hypertension, whereas its activation lowers blood pressure. Evidence suggests that these effects may be attributable to PPARγ activity in the vasculature. However, the specific transcriptional targets of PPARγ in vessels remain largely unidentified. In this study, we examined the role of smooth muscle PPARγ during salt-sensitive hypertension and investigated its transcriptional targets and functional effect. Transgenic mice expressing dominant-negative PPARγ (S-P467L) in smooth muscle cells were more prone to deoxycorticosterone acetate-salt-induced hypertension and mesenteric arterial dysfunction compared with nontransgenic controls. Despite similar morphometry at baseline, vascular remodeling in conduit and small arteries was enhanced in S-P467L after deoxycorticosterone acetate-salt treatment. Gene expression profiling in aorta and mesenteric arteries revealed significantly decreased expression of tissue inhibitor of metalloproteinase-4 (TIMP-4) in S-P467L. Expression of TIMP-4 was increased by deoxycorticosterone acetate-salt treatment, but this increase was ablated in S-P467L. Interference with PPARγ activity either by treatment with a PPARγ inhibitor, GW9662, or by expressing P467L PPARγ markedly suppressed TIMP-4 in primary smooth muscle cells. PPARγ binds to a PPAR response element (PPRE) in chromatin close to the TIMP-4 gene in smooth muscle cells, suggesting that TIMP-4 is a novel target of PPARγ. The interference with PPARγ and decrease in TIMP-4 were accompanied by an increase in total matrix metalloproteinase activity. PPARγ-mediated loss of TIMP-4 increased, whereas overexpression of TIMP-4 decreased smooth muscle cell migration in a scratch assay. Our findings highlight a protective mechanism induced by PPARγ in deoxycorticosterone acetate-salt treatment, establishing a novel mechanistic link between PPARγ and TIMP-4.


Subject(s)
DNA/genetics , Gene Expression Regulation , Hypertension/genetics , Muscle, Smooth, Vascular/metabolism , PPAR gamma/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Animals , Blood Pressure/physiology , Desoxycorticosterone Acetate/toxicity , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/physiopathology , PPAR gamma/metabolism , Tissue Inhibitor of Metalloproteinases/antagonists & inhibitors , Vasoconstriction , Tissue Inhibitor of Metalloproteinase-4
10.
Nat Neurosci ; 17(8): 1083-91, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952644

ABSTRACT

Acid-sensing ion channel 1A (ASIC1A) is abundant in the nucleus accumbens (NAc), a region known for its role in addiction. Because ASIC1A has been suggested to promote associative learning, we hypothesized that disrupting ASIC1A in the NAc would reduce drug-associated learning and memory. However, contrary to this hypothesis, we found that disrupting ASIC1A in the mouse NAc increased cocaine-conditioned place preference, suggesting an unexpected role for ASIC1A in addiction-related behavior. Moreover, overexpressing ASIC1A in rat NAc reduced cocaine self-administration. Investigating the underlying mechanisms, we identified a previously unknown postsynaptic current during neurotransmission that was mediated by ASIC1A and ASIC2 and thus well positioned to regulate synapse structure and function. Consistent with this possibility, disrupting ASIC1A altered dendritic spine density and glutamate receptor function, and increased cocaine-evoked plasticity, which resemble changes previously associated with cocaine-induced behavior. Together, these data suggest that ASIC1A inhibits the plasticity underlying addiction-related behavior and raise the possibility of developing therapies for drug addiction by targeting ASIC-dependent neurotransmission.


Subject(s)
Acid Sensing Ion Channels/physiology , Cocaine/antagonists & inhibitors , Neural Inhibition/genetics , Neuronal Plasticity/genetics , Nucleus Accumbens/physiology , Synaptic Transmission/genetics , Acid Sensing Ion Channels/deficiency , Animals , Behavior, Animal , Cocaine-Related Disorders/metabolism , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neural Inhibition/drug effects , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , Rats , Synaptic Transmission/drug effects , Up-Regulation/genetics
11.
Anat Res Int ; 2011: 580864, 2011.
Article in English | MEDLINE | ID: mdl-22567295

ABSTRACT

The hip region is examined to determine what aspects of musculoskeletal anatomy are precociously developed in primate species with highly specialized modes of locomotion. Muscles of the hind limb were removed and weighed in each specimen, and the hip joint of selected specimens was studied in stained serial sections. No perinatal differences among species are evident, but in adults, the hip joint of Galago moholi (a leaping specialist) appears to have proportionally thick articular cartilage (relative to the subchondral plate) compared to two species of cheirogaleids. Muscle mass distribution in the hind limbs confirms previous observations that the quadriceps femoris muscle is especially large in Galago (in percent mass of the entire hind limb), while the hip region is smaller compared to the more quadrupedal cheirogaleids. Across age groups, the species with the least specialized locomotion as adults, Cheirogaleus medius, shows little or no change in proximal to distal percentage distribution of muscle mass. Galago has a larger percentage mass gain in the thigh. We suggest that muscle mass gain to specific limb segments may be a critical milestone for primates with extremely specialized modes of locomotion.

12.
ASN Neuro ; 1(1)2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19570023

ABSTRACT

Neurological deficits caused by H-I (hypoxia-ischaemia) to the perinatal brain are often severely debilitating and lead to motor impairment, intellectual disability and seizures. Perinatal brain injury is distinct from adult brain injury in that the developing brain is undergoing the normal process of neuronal elimination by apoptotic cell death and thus the apoptotic machinery is more easily engaged and activated in response to injury. Thus cell death in response to neonatal H-I brain injury is partially due to mitochondrial dysfunction and activation of the apoptosome and caspase 3. An important regulator of the apoptotic response following mitochondrial dysfunction is XIAP (X-linked inhibitor of apoptosis protein). XIAP inhibits apoptosis at the level of caspase 9 and caspase 3 activation, and lack of XIAP in vitro has been shown to lead to increased apoptotic cell death. In the present study we show that mice lacking the gene encoding the XIAP protein have an exacerbated response to neonatal H-I injury as measured by tissue loss at 7 days following the injury. In addition, when the XIAP-deficient mice were studied at 24 h post-H-I we found that the increase in injury correlates with an increased apoptotic response in the XIAP-deficient mice and also with brain imaging changes in T2-weighted magnetic resonance imaging and apparent diffusion coefficient that correspond to the location of apoptotic cell death. These results identify a critical role of XIAP in regulating neuronal apoptosis in vivo and demonstrate the enhanced vulnerability of neurons to injury in the absence of XIAP in the developing brain.


Subject(s)
Apoptosis/physiology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain/metabolism , Brain/pathology , X-Linked Inhibitor of Apoptosis Protein/deficiency , Animals , Animals, Newborn , Atrophy , Brain/growth & development , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL