Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
3.
Anticancer Agents Med Chem ; 21(16): 2142-2162, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33563186

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) firstly emerged in Wuhan, China at the end of 2019. After going through the experimental process, the virus was named the novel coronavirus (2019-nCoV) by the World Health Organization (WHO) in February 2020 which has created a global pandemic. The coronavirus disease 2019 (COVID-19) infection is challenging the people who are especially suffering from chronic health problems such as asthma, diabetes, and heart disease or immune system deteriorating disorders, including cancers, Alzheimer's, etc. Other predisposing/risk factors consist of smoking and age (elderly people are at higher risk). The 2019-nCoV attacks epithelial cells in all organs, particularly epithelial cells in the lungs, resulting in viral pneumonia. The 2019-nCoV starts its invasion with the attachment and entry into the respiratory tract epithelial cells via Angiotensin-Converting Enzyme 2 (ACE2) receptors on the epithelial cells. The critical problem with 2019-nCoV is its ability in human to human asymptomatic transmission which causes the rapid and hidden spread of the virus among the population. Also, there are several reports of highly variable and tightly case-dependent clinical manifestations caused by SARS-CoV2, which made the virus more enigmatic. The clinical symptoms are varied from common manifestations which occurred in flu and cold, such as cough, fever, body-ache, trembling, and runny nose to severe conditions, like the Acute Respiratory Distress Syndrome (ARDS) or even uncommon/unusual symptoms such as anosmia, skin color change, and stroke. In fact, besides serious injuries in the respiratory system, COVID-19 invades and damages various organs, including the kidney, liver, gastrointestinal, and nervous system. Accordingly, to cut the transmission chain of disease and control the infection spread. One of the major solutions seems to be early detection of the carriers, particularly the asymptomatic people, with sensitive and accurate diagnostic techniques. Moreover, developing novel and appropriate therapeutic approaches will contribute to the suitable management of the pandemic. Therefore, there is an urgent necessity to make comprehensive investigations and study reviews about COVID-19, offering the latest findings of novel therapies, drugs, epidemiology, and routes of virus transmission and pathogenesis. In this review, we discuss new therapeutic outcomes and cover and the most significant aspects of COVID-19, including the epidemiology, biological features, organs failure, and diagnostic techniques.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , COVID-19 Testing/methods , Adipose Tissue/virology , COVID-19/epidemiology , COVID-19/etiology , COVID-19/pathology , COVID-19/therapy , Female , Humans , Mesenchymal Stem Cell Transplantation , Pregnancy , Pregnancy Complications, Infectious/etiology , Pregnancy Complications, Infectious/virology , Pulmonary Embolism/virology
4.
Mol Cell Endocrinol ; 520: 111095, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33253762

ABSTRACT

The literature has reported a higher prevalence of negative clinical outcomes due to Coronavirus disease 19 (COVID-19) in obese individuals. This can be explained by the cytokine storm, result from the cytokine production from both obesity and viral infection. Gamma-oryzanol (γOz) is a compound with anti-inflammatory and antioxidant activities. However, little is known about the γOz action as a possible agonist of peroxisome proliferator-activated receptor gamma (PPAR-γ). The aim of this study was to test the hypothesis that γOz attenuates the cytokine storm by stimulating PPAR-γ in the adipose tissue. METHODS: Male Wistar rats were randomly divided into three experimental groups and fed ad libitum for 30 weeks with control diet (C, n = 6), high sugar-fat diet (HSF, n = 6) or high sugar-fat diet + Î³Oz (HSF + Î³Oz, n = 6). HSF groups also received water + sucrose (25%). The γOz dose was 0.5% in the chow. Evaluation in animals included caloric intake, body weight, adiposity index, plasma triglycerides, and HOMA-IR. In adipose tissue was evaluated: PPAR-γ gene and protein expression, inflammatory and oxidative stress parameters, and histological analysis. RESULTS: Adipose tissue dysfunction was observed in HSF group, which presented remarkable PPAR-γ underexpression and increased levels of cytokines, other inflammatory markers and oxidative stress. The γOz treatment prevented adipose tissue dysfunction and promoted PPAR-γ overexpression. CONCLUSION: Natural compounds as γOz can be considered a coadjutant therapy to prevent the cytokine storm in COVID-19 patients with obesity conditions.


Subject(s)
Adipose Tissue/metabolism , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Oxidative Stress/drug effects , PPAR gamma/metabolism , Phenylpropionates/pharmacology , SARS-CoV-2/metabolism , Adipose Tissue/pathology , Adipose Tissue/virology , Animals , COVID-19/metabolism , COVID-19/pathology , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/virology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Male , Random Allocation , Rats , Rats, Wistar
5.
Elife ; 92020 09 15.
Article in English | MEDLINE | ID: mdl-32930095

ABSTRACT

Obesity and diabetes are established comorbidities for COVID-19. Adipose tissue demonstrates high expression of ACE2 which SARS- CoV-2 exploits to enter host cells. This makes adipose tissue a reservoir for SARS-CoV-2 viruses and thus increases the integral viral load. Acute viral infection results in ACE2 downregulation. This relative deficiency can lead to disturbances in other systems controlled by ACE2, including the renin-angiotensin system. This will be further increased in the case of pre-conditions with already compromised functioning of these systems, such as in patients with obesity and diabetes. Here, we propose that interactions of virally-induced ACE2 deficiency with obesity and/or diabetes leads to a synergistic further impairment of endothelial and gut barrier function. The appearance of bacteria and/or their products in the lungs of obese and diabetic patients promotes interactions between viral and bacterial pathogens, resulting in a more severe lung injury in COVID-19.


Subject(s)
Coronavirus Infections/microbiology , Diabetes Mellitus/microbiology , Obesity/microbiology , Pneumonia, Viral/microbiology , Adipose Tissue/metabolism , Adipose Tissue/virology , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/isolation & purification , COVID-19 , Comorbidity , Coronavirus Infections/complications , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Diabetes Complications/metabolism , Diabetes Complications/microbiology , Diabetes Complications/virology , Diabetes Mellitus/metabolism , Diabetes Mellitus/virology , Down-Regulation , Host Microbial Interactions , Humans , Microbial Interactions , Obesity/metabolism , Obesity/virology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/complications , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Renin-Angiotensin System , SARS-CoV-2 , Viral Load
6.
Biochimie ; 179: 257-265, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32649962

ABSTRACT

It is becoming obvious that in addition to aging and various hearth pathologies, excess of body weight, especially obesity is a major risk factor for severity of COVID-19 infection. Intriguingly the receptor for SARS-CoV-2 is ACE2, a member of the angiotensin receptor family that has a relatively large tissue distribution. This observation likely explains the multitude of symptoms that have been described from human patients. The adipose tissue also expresses ACE2, suggesting that adipocytes are potentially infected by SARS-CoV-2. Here we discuss some of the potential contribution of the adipose tissue to the severity of the infection and propose some aspects of obese patients metabolic phenotyping to help stratification of individuals with high risk of severe disease.


Subject(s)
COVID-19/complications , Obesity/complications , Adipose Tissue/pathology , Adipose Tissue/virology , Cytokines/metabolism , Humans , Obesity/metabolism , Obesity/pathology , Prevalence
7.
Obesity (Silver Spring) ; 28(7): 1191-1194, 2020 07.
Article in English | MEDLINE | ID: mdl-32314868

ABSTRACT

Coronavirus disease 2019 (COVID-19), the worst pandemic in more than a century, has claimed >125,000 lives worldwide to date. Emerging predictors for poor outcomes include advanced age, male sex, preexisting cardiovascular disease, and risk factors including hypertension, diabetes, and, more recently, obesity. This article posits new obesity-driven predictors of poor COVID-19 outcomes, over and above the more obvious extant risks associated with obesity, including cardiometabolic disease and hypoventilation syndrome in intensive care patients. This article also outlines a theoretical mechanistic framework whereby adipose tissue in individuals with obesity may act as a reservoir for more extensive viral spread, with increased shedding, immune activation, and cytokine amplification. This paper proposes studies to test this reservoir concept with a focus on specific cytokine pathways that might be amplified in individuals with obesity and COVID-19. Finally, this paper underscores emerging therapeutic strategies that might benefit subsets of patients in which cytokine amplification is excessive and potentially fatal.


Subject(s)
Adipose Tissue/virology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Cytokines/metabolism , Obesity/immunology , Pneumonia, Viral/immunology , COVID-19 , Coronavirus Infections/virology , Humans , Lymphocyte Activation/immunology , Obesity/virology , Pandemics , Pneumonia, Viral/virology , SARS-CoV-2
8.
Infez Med ; 27(3): 290-298, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31545773

ABSTRACT

Mediastinal fat has been suggested to be associated with cardiovascular diseases such as carotid stiffness, atherosclerosis and coronary artery calcification. We investigated the possible role of Ad-36-induced obesity in the pathogenesis of the coronary artery disease (CAD). Ad-36 DNA was investigated in the anterior mediastinal fat tissue samples of obese adults with CAD. Seventy-five obese adults with left main coronary artery (LMCA) disease, 28 non-obese adults with valvular heart diseases, and 48 healthy individuals without cardiovascular problems were included as the obese patient group (OPG), non-obese patient group (NOG) and healthy control group (HCG), respectively. We also simultaneously investigated Ad-36 antibodies by serum neutralization test (SNA), and measured leptin and adinopectin levels. Ad-36 antibodies were detected only in 10 patients (13.3%) within the 75 OPG. A statistically significant difference was detected between OPG, NOG and HCG in terms of Ad-36 antibody positivity (p>0.05). Ad-36 DNA was not detected in mediastinal tissue samples of OPG and NOP without PCR inhibitors. We suggest that Ad-36 may not have an affinity for mediastinal adipose tissue in obese patients with left main CAD and valvular heart diseases. Ad-36 antibody positivity results are not sufficient to reach a causal relationship.


Subject(s)
Adenoviruses, Human/immunology , Adipogenesis , Adipose Tissue/virology , Antibodies, Viral/blood , Coronary Artery Disease/etiology , Obesity/virology , Adenoviruses, Human/genetics , Adiponectin/blood , Adult , Case-Control Studies , Coronary Artery Disease/virology , Cross-Sectional Studies , DNA, Viral/isolation & purification , Female , Heart Valve Diseases/virology , Humans , Leptin/blood , Male , Mediastinum/virology , Middle Aged , Obesity/complications , Vascular Calcification , Waist-Hip Ratio
9.
PLoS Pathog ; 15(6): e1007890, 2019 06.
Article in English | MEDLINE | ID: mdl-31220189

ABSTRACT

Cytomegalovirus (CMV) is a ubiquitous herpesvirus infecting most of the world's population. CMV has been rigorously investigated for its impact on lifelong immunity and potential complications arising from lifelong infection. A rigorous adaptive immune response mounts during progression of CMV infection from acute to latent states. CD8 T cells, in large part, drive this response and have very clearly been demonstrated to take up residence in the salivary gland and lungs of infected mice during latency. However, the role of tissue resident CD8 T cells as an ongoing defense mechanism against CMV has not been studied in other anatomical locations. Therefore, we sought to identify additional locations of anti-CMV T cell residency and the physiological consequences of such a response. Through RT-qPCR we found that mouse CMV (mCMV) infected the visceral adipose tissue and that this resulted in an expansion of leukocytes in situ. We further found, through flow cytometry, that adipose tissue became enriched in cytotoxic CD8 T cells that are specific for mCMV antigens from day 7 post infection through the lifespan of an infected animal (> 450 days post infection) and that carry markers of tissue residence. Furthermore, we found that inflammatory cytokines are elevated alongside the expansion of CD8 T cells. Finally, we show a correlation between the inflammatory state of adipose tissue in response to mCMV infection and the development of hyperglycemia in mice. Overall, this study identifies adipose tissue as a location of viral infection leading to a sustained and lifelong adaptive immune response mediated by CD8 T cells that correlates with hyperglycemia. These data potentially provide a mechanistic link between metabolic syndrome and chronic infection.


Subject(s)
Adipose Tissue , CD8-Positive T-Lymphocytes , Herpesviridae Infections , Hyperglycemia , Muromegalovirus/immunology , Panniculitis , Adipose Tissue/immunology , Adipose Tissue/pathology , Adipose Tissue/virology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Hyperglycemia/genetics , Hyperglycemia/immunology , Hyperglycemia/pathology , Hyperglycemia/virology , Immunologic Memory , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Mice , Mice, Knockout , Panniculitis/genetics , Panniculitis/immunology , Panniculitis/pathology , Panniculitis/virology
10.
Nat Immunol ; 20(6): 701-710, 2019 06.
Article in English | MEDLINE | ID: mdl-31110314

ABSTRACT

Cachexia represents a leading cause of morbidity and mortality in various cancers, chronic inflammation and infections. Understanding of the mechanisms that drive cachexia has remained limited, especially for infection-associated cachexia (IAC). In the present paper we describe a model of reversible cachexia in mice with chronic viral infection and identify an essential role for CD8+ T cells in IAC. Cytokines linked to cancer-associated cachexia did not contribute to IAC. Instead, virus-specific CD8+ T cells caused morphologic and molecular changes in the adipose tissue, which led to depletion of lipid stores. These changes occurred at a time point that preceded the peak of the CD8+ T cell response and required T cell-intrinsic type I interferon signaling and antigen-specific priming. Our results link systemic antiviral immune responses to adipose-tissue remodeling and reveal an underappreciated role of CD8+ T cells in IAC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cachexia/etiology , Virus Diseases/complications , Virus Diseases/immunology , Adipose Tissue/diagnostic imaging , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/virology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cachexia/diagnostic imaging , Cachexia/metabolism , Cachexia/pathology , Chronic Disease , Cytokines/blood , Cytokines/metabolism , Female , Interferon Type I/metabolism , Lipid Metabolism , Lipolysis , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus , Magnetic Resonance Imaging/methods , Male , Mice , Signal Transduction , Virus Diseases/virology
11.
Lipids Health Dis ; 18(1): 70, 2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30902099

ABSTRACT

BACKGROUND: This study aims to investigate the molecular mechanism of Adenovirus type 36 (Ad36) in adipocyte differentiation and glucolipid metabolism. METHODS: Rat obesity model was established by Ad36 infection and high-fat diet, respectively. Comparison of the body weight, clinical biochemical indicators, insulin sensitivity and lipid heterotopic deposition between these two models was performed. Ad36-induced adipocyte in vitro model was also established. The binding rate of FoxO1, PPARγ and its target gene promoter was detected using ChIP. The mRNA and protein expression levels of PPARγ and downstream target genes were detected by RT-PCR and Western blot, respectively. Oil red O staining was used to measure differentiation into adipocyte. Wortmannin (WM), inhibitor of PI3K, was used to act on Ad36-induced hADSCs. RESULTS: Ad36-induced obese rats did not exhibit disorders in blood glucose and blood TG, insulin resistance and lipid ectopic deposition. The expression of Adipoq, Lpin1 and Glut4 in the adipose tissue increased. Oil red O staining showed that Ad36 induced the differentiation of hAMSCs into human adipocytes in vitro. During this process, the binding rate of FoxO1 and PPARγ promoter regions was weakened. However, the binding rate of the transcription factor PPARγ to its target genes Acc, Adipoq, Lpin1 and Glut4 was enhanced, and thus increased the protein expression of P-FoxO1, PPARγ2, ACC, LPIN1, GLUT4 and ADIPOQ. The PI3K inhibitor Wortmannin reduced the expression of P-Akt, P-FoxO1 and PPARγ2, thereby inhibiting adipogenesis of hADSC. CONCLUSION: Ad36 may promote fatty acid and triglyceride synthesis, and improve insulin sensitivity by affecting the PI3K/Akt/FoxO1/PPARγ signaling pathway.


Subject(s)
Adipose Tissue/metabolism , Obesity/genetics , PPAR gamma/genetics , Stem Cells/cytology , Adipocytes/metabolism , Adipocytes/virology , Adiponectin/genetics , Adipose Tissue/cytology , Adipose Tissue/virology , Animals , Cell Differentiation/genetics , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Developmental , Glucose/metabolism , Glucose Transporter Type 4/genetics , Humans , Lipid Metabolism/genetics , Obesity/metabolism , Obesity/pathology , Obesity/virology , Phosphatidate Phosphatase/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects , Stem Cells/virology , Wortmannin/administration & dosage
12.
J Infect Dis ; 220(3): 420-431, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30893434

ABSTRACT

Metabolic complications relating to complex effects of viral and immune-mediated mechanisms are now a focus of clinical care among persons living with human immunodeficiency virus (PLHIV), and obesity is emerging as a critical problem. To address knowledge gaps, the US National Institutes of Health sponsored a symposium in May 2018 entitled "Obesity and Fat Metabolism in HIV-infected Individuals." Mechanisms relating to adipose dysfunction and fibrosis, immune function, inflammation, and gastrointestinal integrity were highlighted as contributors to obesity among PLHIV. Fibrotic subcutaneous adipose tissue is metabolically dysfunctional and loses its capacity to expand, leading to fat redistribution, including visceral obesity and ectopic fat accumulation, promoting insulin resistance. Viral proteins, including viral protein R and negative regulatory factor, have effects on adipogenic pathways and cellular metabolism in resident macrophages and T cells. HIV also affects immune cell trafficking into the adipose compartments, with effects on adipogenesis, lipolysis, and ectopic fat accumulation. Key cellular metabolic functions are likely to be affected in PLHIV by gut-derived cytokines and altered microbiota. There are limited strategies to reduce obesity specifically in PLHIV. Enhancing our understanding of critical pathogenic mechanisms will enable the development of novel therapeutics that may normalize adipose tissue function and distribution, reduce inflammation, and improve insulin sensitivity in PLHIV.


Subject(s)
Fats/metabolism , HIV Infections/metabolism , HIV Infections/pathology , Lipid Metabolism/physiology , Obesity/pathology , Obesity/virology , Adipocytes/metabolism , Adipocytes/pathology , Adipocytes/virology , Adipogenesis/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/virology , Adolescent , Adult , Cytokines/metabolism , Female , HIV/pathogenicity , HIV Infections/virology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Insulin Resistance/physiology , Male , Middle Aged , Obesity/metabolism , Viral Proteins/metabolism , Young Adult
13.
J Endocrinol ; 238(1): 61-75, 2018 07.
Article in English | MEDLINE | ID: mdl-29743341

ABSTRACT

The etiopathogenesis of type 1 diabetes (T1D) remains poorly understood. We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to better understand the role of the innate immune system in the mechanism of virus-induced disease. We observed that infection with KRV results in cell influx into visceral adipose tissue soon following infection prior to insulitis and hyperglycemia. In sharp contrast, subcutaneous adipose tissue is free of cellular infiltration, whereas ß cell inflammation and diabetes are observed beginning on day 14 post infection. Immunofluorescence studies further demonstrate that KRV triggers CD68+ macrophage recruitment and the expression of KRV transcripts and proinflammatory cytokines and chemokines in visceral adipose tissue. Adipocytes from naive rats cultured in the presence of KRV express virus transcripts and upregulate cytokine and chemokine gene expression. KRV induces apoptosis in visceral adipose tissue in vivo, which is reflected by positive TUNEL staining and the expression of cleaved caspase-3. Moreover, KRV leads to an oxidative stress response and downregulates the expression of adipokines and genes associated with mediating insulin signaling. Activation of innate immunity with Poly I:C in the absence of KRV leads to CD68+ macrophage recruitment to visceral adipose tissue and a decrease in adipokine expression detected 5 days following Poly (I:C) treatment. Finally, proof-of-principle studies show that brief anti-inflammatory steroid therapy suppresses visceral adipose tissue inflammation and protects from virus-induced disease. Our studies provide evidence raising the hypothesis that visceral adipose tissue inflammation and dysfunction may be involved in early mechanisms triggering ß cell autoimmunity.


Subject(s)
Adipose Tissue/pathology , Adipose Tissue/physiopathology , Diabetes Mellitus, Type 1/virology , Inflammation/complications , Panniculitis/complications , Parvovirus/physiology , Adipose Tissue/immunology , Adipose Tissue/virology , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Female , Immunity, Innate/physiology , Inflammation/pathology , Inflammation/virology , Macrophages/physiology , Male , Panniculitis/immunology , Panniculitis/pathology , Panniculitis/virology , Parvovirus/immunology , Rats , Signal Transduction/immunology
14.
Curr HIV/AIDS Rep ; 15(1): 60-71, 2018 02.
Article in English | MEDLINE | ID: mdl-29423731

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to examine the evidence describing adipose tissue as a reservoir for HIV-1 and how this often expansive anatomic compartment contributes to HIV persistence. RECENT FINDINGS: Memory CD4 T cells and macrophages, the major host cells for HIV, accumulate in adipose tissue during HIV/SIV infection of humans and rhesus macaques. Whereas HIV and SIV proviral DNA is detectable in CD4 T cells of multiple fat depots in virtually all infected humans and monkeys examined, viral RNA is less frequently detected, and infected macrophages may be less prevalent in adipose tissue. However, based on viral outgrowth assays, adipose-resident CD4 T cells are latently infected with virus that is replication-competent and infectious. Additionally, adipocytes interact with CD4 T cells and macrophages to promote immune cell activation and inflammation which may be supportive for HIV persistence. Antiviral effector cells, such as CD8 T cells and NK/NKT cells, are abundant in adipose tissue during HIV/SIV infection and typically exceed CD4 T cells, whereas B cells are largely absent from adipose tissue of humans and monkeys. Additionally, CD8 T cells in adipose tissue of HIV patients are activated and have a late differentiated phenotype, with unique TCR clonotypes of less diversity relative to blood CD8 T cells. With respect to the distribution of antiretroviral drugs in adipose tissue, data is limited, but there may be class-specific penetration of fat depots. The trafficking of infected immune cells within adipose tissues is a common event during HIV/SIV infection of humans and monkeys, but the virus may be mostly transcriptionally dormant. Viral replication may occur less in adipose tissue compared to other major reservoirs, such as lymphoid tissue, but replication competence and infectiousness of adipose latent virus are comparable to other tissues. Due to the ubiquitous nature of adipose tissue, inflammatory interactions among adipocytes and CD4 T cells and macrophages, and selective distribution of antiretroviral drugs, the sequestration of infected immune cells within fat depots likely represents a major challenge for cure efforts.


Subject(s)
Adipocytes/virology , Adipose Tissue/virology , CD4-Positive T-Lymphocytes/virology , Macrophages/virology , Viral Load , Virus Replication/physiology , Animals , Anti-Retroviral Agents/therapeutic use , Antiviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , HIV-1/growth & development , HIV-1/immunology , Humans , Macaca mulatta , Macrophages/immunology , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/immunology
15.
J Infect Dis ; 217(11): 1770-1781, 2018 05 05.
Article in English | MEDLINE | ID: mdl-29401318

ABSTRACT

Background: Fibrosis in lymph nodes may limit CD4+ T-cell recovery, and lymph node and adipose tissue fibrosis may contribute to inflammation and comorbidities despite antiretroviral therapy (ART). We hypothesized that the angiotensin receptor blocker and peroxisome proliferator-activated receptor γ agonist telmisartan would decrease lymph node or adipose tissue fibrosis in treated human immunodeficiency virus type 1 (HIV) infection. Methods: In this 48-week, randomized, controlled trial, adults continued HIV-suppressive ART and received telmisartan or no drug. Collagen I, fibronectin, and phosphorylated SMAD3 (pSMAD3) deposition in lymph nodes, as well as collagen I, collagen VI, and fibronectin deposition in adipose tissue, were quantified by immunohistochemical analysis at weeks 0 and 48. Two-sided rank sum and signed rank tests compared changes over 48 weeks. Results: Forty-four participants enrolled; 35 had paired adipose tissue specimens, and 29 had paired lymph node specimens. The median change overall in the percentage of the area throughout which collagen I was deposited was -2.6 percentage points (P = 0.08) in lymph node specimens and -1.3 percentage points (P = .001) in adipose tissue specimens, with no between-arm differences. In lymph node specimens, pSMAD3 deposition changed by -0.5 percentage points overall (P = .04), with no between-arm differences. Telmisartan attenuated increases in fibronectin deposition (P = .06). In adipose tissue, changes in collagen VI deposition (-1.0 percentage point; P = .001) and fibronectin deposition (-2.4 percentage points; P < .001) were observed, with no between-arm differences. Conclusions: In adults with treated HIV infection, lymph node and adipose tissue fibrosis decreased with continued ART alone, with no additional fibrosis reduction with telmisartan therapy.


Subject(s)
Adipose Tissue/drug effects , Antihypertensive Agents/therapeutic use , Fibrosis/drug therapy , Lymph Nodes/drug effects , Telmisartan/therapeutic use , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/virology , Adult , Antiretroviral Therapy, Highly Active/methods , Female , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/virology , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Middle Aged , PPAR gamma/metabolism
16.
Virus Res ; 244: 90-98, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29141203

ABSTRACT

Human adenovirus (HAdV) type 36 seropositivity has been linked to obesity in humans. That link is supported by a small number of studies using HAdV-36 infection of animals that are not natural hosts for HAdVs. In this study, we infected mice with mouse adenovirus type 1 (MAV-1), a mouse pathogen, to determine whether MAV-1 infected adipose tissue and was associated with adipose tissue inflammation and obesity. We detected MAV-1 in adipose tissue during acute MAV-1 infection, but we did not detect virus-induced increases in adipose tissue cytokine expression or histological evidence of adipose tissue inflammation during acute infection. MAV-1 did not persist in adipose tissue at later times, and we did not detect long-term adipose inflammation, increased adipose tissue mass, or body weight in infected mice. Our data indicate that MAV-1 is not associated with obesity in infected mice.


Subject(s)
Adenoviridae Infections/virology , Adipose Tissue/virology , DNA, Viral/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions , Mastadenovirus/genetics , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Adipose Tissue/immunology , Animals , Body Weight , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/immunology , DNA, Viral/immunology , Female , Fibroblasts/virology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mastadenovirus/growth & development , Mastadenovirus/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virus Replication
17.
Rev Med Chil ; 145(8): 1054-1059, 2017 Aug.
Article in Spanish | MEDLINE | ID: mdl-29189864

ABSTRACT

The evidence of the last 20 years shows a link between viral infections and obesity in animals and humans. There are five adenovirus which have been associated with development of obesity in animals. SMAM-1 virus was the first studied in humans associated with obesity. There is compelling evidence that Ad-36 virus could contribute to the development of obesity in humans and it is related with body mass index (BMI). This manuscript reviews the association between Ad-36 and the other four virus infections with obesity. An electronic search of articles in the databases PubMed and Scielo, with use of key words: obesity, infection, adipose tissue, Ad-36, 3T3-L1 was performed. The search was restricted "human" and "animals". The importance of the relationship between virus infections and obesity has increased over the past two decades. Ad-36 shows more compelling evidence in humans. There are reports involving this virus in the enhancement of adipogenesis, adipocyte differentiation, a lower secretion of leptin and an increased insulin sensitivity. Future work should focus in larger cohort studies to confirm this association, which explains the global obesity epidemic from a new perspective.


Subject(s)
Adenoviridae Infections/complications , Adenoviridae/pathogenicity , Obesity/virology , Adipose Tissue/virology , Animals , Body Mass Index , Humans , Risk Factors
18.
Eur J Immunol ; 47(12): 2113-2123, 2017 12.
Article in English | MEDLINE | ID: mdl-28762530

ABSTRACT

We and others have demonstrated that adipose tissue is a reservoir for HIV. Evaluation of the mechanisms responsible for viral persistence may lead to ways of reducing these reservoirs. Here, we evaluated the immune characteristics of adipose tissue in HIV-infected patients receiving antiretroviral therapy (ART) and in non-HIV-infected patients. We notably sought to determine whether adipose tissue's intrinsic properties and/or HIV induced alteration of the tissue environment may favour viral persistence. ART-controlled HIV infection was associated with a difference in the CD4/CD8 T-cell ratio and an elevated proportion of Treg cells in subcutaneous adipose tissue. No changes in Th1, Th2 and Th17 cell proportions or activation markers expression on T cell (Ki-67, HLA-DR) could be detected, and the percentage of CD69-expressing resident memory CD4+ T cells was not affected. Overall, our results indicate that adipose-tissue-resident CD4+ T cells are not extensively activated during HIV infection. PD-1 was expressed by a high proportion of tissue-resident memory CD4+ T cells in both HIV-infected patients and non-HIV-infected patients. Our findings suggest that adipose tissue's intrinsic immunomodulatory properties may limit immune activation and thus may strongly contribute to viral persistence.


Subject(s)
Adipose Tissue/immunology , CD4-Positive T-Lymphocytes/immunology , Cellular Microenvironment/immunology , HIV Infections/immunology , HIV-1/immunology , Programmed Cell Death 1 Receptor/immunology , Adipose Tissue/metabolism , Adipose Tissue/virology , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Disease Reservoirs/virology , Female , Flow Cytometry , HIV Infections/metabolism , HIV Infections/virology , HIV-1/drug effects , Host-Pathogen Interactions/immunology , Humans , Male , Middle Aged , Programmed Cell Death 1 Receptor/metabolism
19.
Rev. méd. Chile ; 145(8): 1054-1059, ago. 2017.
Article in Spanish | LILACS | ID: biblio-902584

ABSTRACT

The evidence of the last 20 years shows a link between viral infections and obesity in animals and humans. There are five adenovirus which have been associated with development of obesity in animals. SMAM-1 virus was the first studied in humans associated with obesity. There is compelling evidence that Ad-36 virus could contribute to the development of obesity in humans and it is related with body mass index (BMI). This manuscript reviews the association between Ad-36 and the other four virus infections with obesity. An electronic search of articles in the databases PubMed and Scielo, with use of key words: obesity, infection, adipose tissue, Ad-36, 3T3-L1 was performed. The search was restricted "human" and "animals". The importance of the relationship between virus infections and obesity has increased over the past two decades. Ad-36 shows more compelling evidence in humans. There are reports involving this virus in the enhancement of adipogenesis, adipocyte differentiation, a lower secretion of leptin and an increased insulin sensitivity. Future work should focus in larger cohort studies to confirm this association, which explains the global obesity epidemic from a new perspective.


Subject(s)
Humans , Animals , Adenoviridae/pathogenicity , Adenoviridae Infections/complications , Obesity/virology , Body Mass Index , Adipose Tissue/virology , Risk Factors
20.
J Microbiol ; 55(1): 75-79, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28035603

ABSTRACT

Naturally occurring reoviruses are live replication-proficient viruses specifically infecting human cancer cells while sparing the normal counterparts. Stem cells can be highly susceptible to viral infection due to their innate high proliferation potential and other active signaling pathways of cells that might be involved in viral tropism. In the previous study, we showed that reoviruses could adversely affect murine embryonic stem cells' integrity in vitro and in vivo. Oncolytic viruses, delivered systemically face many hurdles that also impede their localization and infection of, metastatic tumors, due to a variety of immune and physical barriers. To overcome such hurdles to systemic delivery, several studies supported the idea that certain types of cells, including mesenchymal stem cells, might play a role as cell carriers for oncolytic viruses. Thus, it would be interesting to examine whether human adult stem cells such as human adipose-derived mesenchymal stem cells could be saved by the reoviral challenge. In this study, we report that biological activities such as proliferation and multipotency of human adipose-derived stem cells are not affected by wild-type reovirus challenge as evidenced by survival, osteogenic and adipogenic differentiation potential assays following treatment with reoviruses. Therefore, unlike murine embryonic stem cells, our study strongly suggests that human adipose-derived adult stem cells could be spared in vivo during wild-type reoviral anti-cancer therapeutics in a clinical setting. Furthermore, the results support the possible clinical use of human adipose-derived stem cells as an effective cell carrier of oncolytic reovirus to maximize their tumor tropism and anti-tumor activity.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/virology , Mesenchymal Stem Cells/physiology , Mesenchymal Stem Cells/virology , Reoviridae/physiology , Adipogenesis , Adipose Tissue/physiology , Adipose Tissue/virology , Adult Stem Cells/physiology , Adult Stem Cells/ultrastructure , Cell Proliferation , Humans , Mesenchymal Stem Cells/ultrastructure , Neoplasms/therapy , Osteogenesis , Viral Tropism/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...