Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Front Immunol ; 12: 744782, 2021.
Article in English | MEDLINE | ID: mdl-34721414

ABSTRACT

Introduction: There is evidence that obesity, a risk factor for asthma severity and morbidity, has a unique asthma phenotype which is less atopic and less responsive to inhaled corticosteroids (ICS). Peripheral blood mononuclear cells (PBMC) are important to the immunologic pathways of obese asthma and steroid resistance. However, the cellular source associated with steroid resistance has remained elusive. We compared the lymphocyte landscape among obese children with asthma to matched normal weight children with asthma and assessed relationship to asthma control. Methods: High-dimensional flow cytometry of PBMC at baseline and after dexamethasone stimulation was performed to characterize lymphocyte subpopulations, T-lymphocyte polarization, proliferation (Ki-67+), and expression of the steroid-responsive protein FK506-binding protein 51 (FKBP51). T-lymphocyte populations were compared between obese and normal-weight participants, and an unbiased, unsupervised clustering analysis was performed. Differentially expressed clusters were compared with asthma control, adjusted for ICS and exhaled nitric oxide. Results: In the obese population, there was an increased cluster of CD4+ T-lymphocytes expressing Ki-67 and FKBP51 at baseline and CD4+ T-lymphocytes expressing FKBP51 after dexamethasone stimulation. CD4+ Ki-67 and FKBP51 expression at baseline showed no association with asthma control. Dexamethasone-induced CD4+ FKBP51 expression was associated with worse asthma control in obese participants with asthma. FKBP51 expression in CD8+ T cells and CD19+ B cells did not differ among groups, nor did polarization profiles for Th1, Th2, Th9, or Th17 percentage. Discussion: Dexamethasone-induced CD4+ FKBP51 expression is uniquely associated with worse asthma control in obese children with asthma and may underlie the corticosteroid resistance observed in this population.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , CD4-Positive T-Lymphocytes/immunology , Dexamethasone/therapeutic use , Pediatric Obesity/complications , Tacrolimus Binding Proteins/biosynthesis , Air Filters , Asthma/complications , Asthma/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Child , Drug Resistance/immunology , Female , Humans , Male , Particulate Matter/adverse effects , Pediatric Obesity/immunology
2.
J Pediatr ; 239: 155-160, 2021 12.
Article in English | MEDLINE | ID: mdl-34454951

ABSTRACT

OBJECTIVES: To evaluate the prevalence and clinical significance of autoantibodies in children with overweight and obesity with nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH) compared with those with autoimmune liver disease (ALD). STUDY DESIGN: This was a retrospective, cross-sectional study of children with a biopsy-proven diagnosis of NAFL, NASH, autoimmune hepatitis (AIH), or primary sclerosing cholangitis (PSC) and a body mass index (BMI) >85th percentile treated between 2007 and 2016. RESULTS: A total of 181 patients were identified, including 31 (17%) with NAFL, 121 (67%) with NASH, 12 (6.6%) with ALD (AIH, PSC, or overlap), and 17 (9.4%) with combined ALD and NAFLD. Antinuclear antibody (ANA), anti-actin antibody, and anti-liver kidney microsomal (LKM) antibody were positive in 16.1%, 13.8%, and 0%, respectively, of the patients with NAFL and in 32.8%, 15.5%, and 0%, respectively, of those with NASH. Total immunoglobulin G (IgG) was elevated in 27.3% of the patients with NAFL and in 47.7% of those with NASH, but in 100% of those with ALD. The positive predictive value of LKM was 100% for ALD but only 29% for ANA and 46% for anti-actin antibody. CONCLUSIONS: False-positive rates of autoantibodies were higher in pediatric patients with overweight and obesity with NAFLD compared with the general adult population. Positive LKM had the highest specificity and positive predictive value, and elevated IgG level had the highest sensitivity for ALD. The presence of autoantibodies does not signal more severe NAFLD in children. BMI >98th percentile seems to be an important breakpoint above which ALD is less likely.


Subject(s)
Autoantibodies/blood , Cholangitis, Sclerosing/diagnosis , Hepatitis, Autoimmune/diagnosis , Non-alcoholic Fatty Liver Disease/diagnosis , Pediatric Obesity/complications , Adolescent , Biomarkers/blood , Case-Control Studies , Child , Child, Preschool , Cholangitis, Sclerosing/blood , Cholangitis, Sclerosing/complications , Cholangitis, Sclerosing/immunology , Clinical Decision Rules , Cross-Sectional Studies , Diagnosis, Differential , False Positive Reactions , Female , Hepatitis, Autoimmune/blood , Hepatitis, Autoimmune/complications , Hepatitis, Autoimmune/immunology , Humans , Male , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/immunology , Patient Acuity , Pediatric Obesity/blood , Pediatric Obesity/immunology , Retrospective Studies , Sensitivity and Specificity , Young Adult
3.
J Clin Endocrinol Metab ; 106(10): e3923-e3935, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34128988

ABSTRACT

CONTEXT: Adult obesity is associated with chronic low-grade inflammation and may give rise to future chronic disease. However, it is unclear whether adiposity-related inflammation is already apparent in childhood. OBJECTIVE: To study associations between child adiposity measures with circulating monocytes and naive and memory subsets in CD4, CD8, and γδ T cell lineages. METHODS: Ten-year-old children (n = 890) from the Generation R Cohort underwent dual-energy x-ray absorptiometry and magnetic resonance imaging for body composition (body mass index [BMI], fat mass index [FMI], android-to-gynoid fat mass ratio, visceral fat index, liver fat fraction). Blood samples were taken for detailed immunophenotyping of leukocytes by 11-color flow cytometry. RESULTS: Several statistically significant associations were observed. A 1-SD increase in total FMI was associated with +8.4% (95% CI 2.0, 15.2) Vδ2+Vγ9+ and +7.4% (95% CI 2.4, 12.5) CD8+TEMRO cell numbers. A 1-SD increase in visceral fat index was associated with +10.7% (95% CI 3.3, 18.7) Vδ2+Vγ9+ and +8.3% (95% CI 2.6, 14.4) CD8+TEMRO cell numbers. Higher android-to-gynoid fat mass ratio was only associated with higher Vδ2+Vγ9+ T cells. Liver fat was associated with higher CD8+TEMRO cells but not with Vδ2+Vγ9+ T cells. Only liver fat was associated with lower Th17 cell numbers: a 1-SD increase was associated with -8.9% (95% CI -13.7, -3.7) Th17 cells. No associations for total CD8+, CD4+ T cells, or monocytes were observed. BMI was not associated with immune cells. CONCLUSION: Higher Vδ2+Vγ9+ and CD8+TEMRO cell numbers in children with higher visceral fat index could reflect presence of adiposity-related inflammation in children with adiposity of a general population.


Subject(s)
Adiposity/physiology , CD8-Positive T-Lymphocytes/pathology , Pediatric Obesity/immunology , CD8-Positive T-Lymphocytes/metabolism , Child , Cohort Studies , Cross-Sectional Studies , Female , Genes, T-Cell Receptor delta , Genes, T-Cell Receptor gamma , Humans , Immunologic Memory/immunology , Immunophenotyping , Lymphocyte Count , Lymphocyte Subsets/metabolism , Lymphocyte Subsets/pathology , Male , Monocytes/pathology , Netherlands/epidemiology , Pediatric Obesity/blood , Pediatric Obesity/epidemiology
4.
Parasitol Int ; 83: 102343, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33831579

ABSTRACT

Background Toxoplasmosis as a global disease is considered as a triggering factor responsible for development of several clinical diseases. However, Toxoplasma gondii (T. gondii) is an understudied parasite of potential interest in obesity research. The current study aimed to explore the role of latent T. gondii infection in the pathogenesis of metabolic syndrome (MetS) in obese adolescents through studying the relationship between serum interferon-gamma [IFN-γ] and serum chemerin in context of MetS components. Methods Eighty-three obese adolescents were serologically screened for T. gondii-IgG antibodies and compared to 35 age-matched healthy T. gondii-seronegative controls. Participants were evaluated for anthropometric measurements, total-fat mass [FM], trunk-FM, serum lipid profile, IFN-γ, and chemerin levels. Homeostatic Model Assessment of insulin resistance (HOMA-IR) was calculated. Results The prevalence of MetS was significantly higher within obese T. gondii-seropositive group compared to obese T. gondii-seronegative group (P = 0.033). Seropositive obese MetS group displayed significantly higher trunk-FM, HOMA-IR, chemerin, and IFN-γ compared to seronegative obese MetS group. Serum chemerin and IFN-γ were strongly correlated (P < 0.001) and were positively correlated with BMI, WC, total-FM, trunk-FM, HOMA-IR, cholesterol, triglycerides and negatively correlated with HDLC. HOMA-IR was a common predictor for serum chemerin (P = 0.030) and IFN-γ (P < 0.001). Conclusions The study results suggest that T. gondii infection may exert an immune-metabolic effect that may have a potential role in the development of MetS among obese adolescents.


Subject(s)
Metabolic Syndrome/parasitology , Pediatric Obesity/etiology , Toxoplasmosis/complications , Adolescent , Cross-Sectional Studies , Egypt , Female , Humans , Male , Metabolic Syndrome/immunology , Pediatric Obesity/immunology , Toxoplasmosis/immunology
5.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572203

ABSTRACT

Overweight and obesity during pregnancy have been associated with increased birth weight, childhood obesity, and noncommunicable diseases in the offspring, leading to a vicious transgenerational perpetuating of metabolic derangements. Key components in intrauterine developmental programming still remain to be identified. Obesity involves chronic low-grade systemic inflammation that, in addition to physiological adaptations to pregnancy, may potentially expand to the placental interface and lead to intrauterine derangements with a threshold effect. Animal models, where maternal inflammation is mimicked by single injections with lipopolysaccharide (LPS) resembling the obesity-induced immune profile, showed increased adiposity and impaired metabolic homeostasis in the offspring, similar to the phenotype observed after exposure to maternal obesity. Cytokine levels might be specifically important for the metabolic imprinting, as cytokines are transferable from maternal to fetal circulation and have the capability to modulate placental nutrient transfer. Maternal inflammation may induce metabolic reprogramming at several levels, starting from the periconceptional period with effects on the oocyte going through early stages of embryonic and placental development. Given the potential to reduce inflammation through inexpensive, widely available therapies, examinations of the impact of chronic inflammation on reproductive and pregnancy outcomes, as well as preventive interventions, are now needed.


Subject(s)
Child Development/physiology , Fetal Development/immunology , Obesity, Maternal/immunology , Pediatric Obesity/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Child , Disease Models, Animal , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Maternal Nutritional Physiological Phenomena/immunology , Maternal-Fetal Exchange/immunology , Metabolic Networks and Pathways/immunology , Obesity, Maternal/complications , Obesity, Maternal/metabolism , Obesity, Maternal/therapy , Pediatric Obesity/metabolism , Pediatric Obesity/prevention & control , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/prevention & control
6.
Diabetologia ; 64(4): 826-835, 2021 04.
Article in English | MEDLINE | ID: mdl-33474583

ABSTRACT

AIMS/HYPOTHESIS: We aimed to evaluate the relationship between childhood growth measures and risk of developing islet autoimmunity (IA) and type 1 diabetes in children with an affected first-degree relative and increased HLA-conferred risk. We hypothesised that being overweight or obese during childhood is associated with a greater risk of IA and type 1 diabetes. METHODS: Participants in a randomised infant feeding trial (N = 2149) were measured at 12 month intervals for weight and length/height and followed for IA (at least one positive out of insulin autoantibodies, islet antigen-2 autoantibody, GAD autoantibody and zinc transporter 8 autoantibody) and development of type 1 diabetes from birth to 10-14 years. In this secondary analysis, Cox proportional hazard regression models were adjusted for birthweight and length z score, sex, HLA risk, maternal type 1 diabetes, mode of delivery and breastfeeding duration, and stratified by residence region (Australia, Canada, Northern Europe, Southern Europe, Central Europe and the USA). Longitudinal exposures were studied both by time-varying Cox proportional hazard regression and by joint modelling. Multiple testing was considered using family-wise error rate at 0.05. RESULTS: In the Trial to Reduce IDDM in the Genetically at Risk (TRIGR) population, 305 (14.2%) developed IA and 172 (8%) developed type 1 diabetes. The proportions of children overweight (including obese) and obese only were 28% and 9% at 10 years, respectively. Annual growth measures were not associated with IA, but being overweight at 2-10 years of life was associated with a twofold increase in the development of type 1 diabetes (HR 2.39; 95% CI 1.46, 3.92; p < 0.001 in time-varying Cox regression), and similarly with joint modelling. CONCLUSIONS/INTERPRETATION: In children at genetic risk of type 1 diabetes, being overweight at 2-10 years of age is associated with increased risk of progression from multiple IA to type 1 diabetes and with development of type 1 diabetes, but not with development of IA. Future studies should assess the impact of weight management strategies on these outcomes. TRIAL REGISTRATION: ClinicalTrials.gov NCT00179777.


Subject(s)
Adolescent Development , Autoimmunity/genetics , Child Development , Diabetes Mellitus, Type 1/epidemiology , Islets of Langerhans/immunology , Pediatric Obesity/epidemiology , Adolescent , Age Factors , Australia/epidemiology , Bottle Feeding , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Europe/epidemiology , Female , Genetic Predisposition to Disease , Heredity , Humans , Incidence , Infant , Infant Formula , Infant, Newborn , Male , North America/epidemiology , Pediatric Obesity/immunology , Pediatric Obesity/prevention & control , Pedigree , Phenotype , Prognosis , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors
7.
Pediatr Obes ; 16(1): e12696, 2021 01.
Article in English | MEDLINE | ID: mdl-32638543

ABSTRACT

BACKGROUND: Autoantibodies are frequently positive in adults with nonalcoholic fatty liver disease (NAFLD) without concurrent autoimmune hepatitis (AIH). The clinical significance of this is unknown in children. OBJECTIVE: To determine the prevalence of autoantibody positivity in pediatric NAFLD and to evaluate its association with disease severity. METHODS: Multicenter, retrospective study of patients ≤18 years of age with biopsy-confirmed NAFLD. Descriptive statistics were used and groups were compared using Wilcoxon-Mann Whitney or χ2 testing, and multivariable logistic regression was used for binary or ordinal outcomes. RESULTS: One hundred and thirty six patients with a median age of 14 years were included. The median body mass index Z-score was 2.5 (interquartile range 2.2, 2.6). Positive antinuclear antibody (ANA), anti-smooth muscle antibody (ASMA), anti-liver-kidney microsomal antibody, or any combination of autoantibodies were observed in 22%, 14%, 0%, and 33% of patients, respectively. The proportion of patients with a steatosis score ≥2 was significantly higher in those with positive ANA (P = .045). In the multivariable regression analysis, positive ANA was associated with increased odds of steatosis score ≥2 (odds ratio, 5.91; 95% confidential interval, 1.50-23.26), after controlling for potential confounders. No other significant histology differences were seen between the groups. CONCLUSIONS: Positive ANA and ASMA are common in children with NAFLD; however, anti-LKM positivity is not. ANA positivity is associated with more severe steatosis.


Subject(s)
Autoantibodies/blood , Non-alcoholic Fatty Liver Disease/immunology , Pediatric Obesity/immunology , Adolescent , Biomarkers/blood , Child , Child, Preschool , Female , Humans , Logistic Models , Male , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Pediatric Obesity/blood , Pediatric Obesity/complications , Retrospective Studies , Severity of Illness Index
8.
Curr Opin Pediatr ; 32(6): 805-815, 2020 12.
Article in English | MEDLINE | ID: mdl-33105275

ABSTRACT

PURPOSE OF REVIEW: Childhood obesity, with persistent chronic inflammation, is a worldwide epidemic. Obesity causes dysregulation throughout the immune system, affecting the balance and levels of cytokines, adipokines, and innate and adaptive immune cells. The present review focuses on the impact of obesity on immune function in children: altering the baseline activation state of immune cells and affecting the ability of the host to combat pathogens and malignancy and respond appropriately to vaccination. RECENT FINDINGS: Obesity causes dysregulation of the immune system. Single-cell RNA-sequencing of adipose tissue and resident immune cells is quantifying the impact of obesity on the frequency of immune cell subsets and their states. The system-wide alterations in immune function in obesity are most evident upon perturbation, including the response to infection (e.g. increased risk of severe COVID-19 in the ongoing pandemic), vaccination, and malignancy. However, mechanistic research in pediatric obesity is limited and this impacts our ability to care for these children. SUMMARY: We must better understand baseline and perturbed immune health in obese children to determine how to account for altered frequency and function of humoral and cellular immune components in acute infection, during vaccine design and when considering therapeutic options for this complex, medically vulnerable group.


Subject(s)
Immune System/physiology , Pediatric Obesity/immunology , Adipokines/immunology , Adipose Tissue/immunology , Child , Cytokines/immunology , Humans , Immunity, Cellular , Immunity, Humoral , Infections/immunology , Vaccination
9.
Curr Obes Rep ; 9(3): 204-212, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32519271

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to summarize our current understanding of the association between childhood obesity and cancer risk later in life. RECENT FINDINGS: Adipose tissue secrets a variety of adipocytokines, and expression and/or secretion rate of most of them seems to be increased or dysregulated in obesity. In addition, obesity leads to increased secretion of proinflammatory cytokines such as interferon-γ (IFN-γ), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α), which promotes an infiltration of inflammatory immune cells into adipose tissue. This process may facilitate a state of "subclinical inflammation" (metaflammation) and may lead to the development of the metabolic syndrome (MetS), starting as early as during childhood. In addition, several oncogenes have been linked to inflammation and cancer development via different pathways, and several types of tumors need an inflammatory environment before a malignant change occurs. An inflammatory environment seems to promote the proliferation and survival of malignant cells as well as angiogenesis. Natural killer (NK) cells play an important role in this process, as they are able to kill transformed cells without prior sensitization and coordinate subsequent immune responses by producing distinct cytokines, thus providing antitumor immunity. First studies in children have suggested that NK cells from obese children are activated, metabolically stressed, and functionally deficient. This may lead to a suppression of antitumor immunity as early as during childhood, probably many years before the development of cancer. Epidemiological studies have shown a strong association between higher body mass index (BMI) during childhood and adolescence and increased risk for several malignancies in adulthood, including leukemia, Hodgkin's disease, colorectal cancer, and breast cancer. Underlying mechanisms are not completely understood, but several adipocytokines and inflammatory markers including NK cells seem to be "key players" in this process.


Subject(s)
Adipose Tissue/immunology , Cytokines/metabolism , Killer Cells, Natural/immunology , Neoplasms/immunology , Pediatric Obesity/immunology , Adiposity/immunology , Adolescent , Adult , Carcinogenesis/immunology , Child , Female , Humans , Inflammation , Male , Neoplasms/etiology , Pediatric Obesity/complications , Risk Factors
10.
Ann Allergy Asthma Immunol ; 125(4): 425-432.e4, 2020 10.
Article in English | MEDLINE | ID: mdl-32561508

ABSTRACT

BACKGROUND: Asthma is a chronic inflammatory condition characterized by T-helper (TH) 2 polarization. In children, the prevalence of obesity is associated with an increased incidence of asthma. Notably, obesity is linked with TH1-mediated inflammation and has been identified as a major risk factor for asthma. OBJECTIVE: To investigate the impact of obesity on TH1 (tumor necrosis factor α, interferon gamma, interleukin (IL)-6, IL-8) and TH2 (IL-4, IL-5, IL-10, IL-13) immune responses in children with asthma. METHODS: We searched the MEDLINE and gray literature electronic databases for eligible studies from inception up until April 2020. The quality of included studies and evidence was independently assessed by 2 reviewers. The random-effects model was used in this meta-analysis, and outcomes were reported as standardized mean difference (SMD) and 95% confidence interval (CI). RESULTS: Overall, 5 studies comprising 482 participants met the inclusion criteria. The meta-analysis revealed an increased TH2-mediated immune response in lean people with asthma compared with controls without asthma (SMD: -1.15 [95% CI: -1.93, 0.36]; I2 = 93%; pH < .001). However, in obese people with asthma, there was polarization toward TH1 immune response compared with lean people with asthma (SMD: -0.43 [95% CI: -0.79, -0.08]; I2 = 88%, pH < .001). CONCLUSION: This meta-analysis reveals that there are differences in immune responses mediated by T-helper cells in lean and obese children with asthma. Moreover, and not unique to asthma, obesity polarizes the immune response toward TH1 rather than the classical TH2. This could be an important aspect to understand to establish effective therapeutic targets for obese children with asthma.


Subject(s)
Asthma/immunology , Pediatric Obesity/complications , Pediatric Obesity/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Child , Female , Humans , Male
11.
Nutr Metab Cardiovasc Dis ; 30(7): 1188-1195, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32448719

ABSTRACT

BACKGROUND AND AIMS: Glomerular hyperfiltration (GH) is proposed as one of the earliest events in obesity (OB)-associated renal disease. Children with GH and type-1 diabetes showed increased chemokine levels. Chemokine associations with glomerular filtration rate (GFR) and metabolic features in prepubertal children with overweight (OW)/OB are unknown. METHODS AND RESULTS: Cross-sectional study. 75 prepubertal children (aged: 9.0 ± 1.7 years) with OW/OB were studied. Clinical and metabolic characteristics (including non-esterified fatty acids, NEFA) and GFR (combined Zappitelli equation) were assessed. GH was defined as GFR >135 ml/min.1.73 m2. Serum levels of regulated on activation, normal T cell expressed and secreted (RANTES)/CCL5, interleukin-8 (IL-8)/CXCL8 and monokine-induced by interferon-γ (MIG)/CXCL9 were measured by ELISA. Age- and sex-adjusted correlations and differences were tested. 48% of the cohort was female and 13% were OW, 54% OB and 33% severe OB. Prepubertal children with GH showed lower z-BMI (-12%), NEFA (-26%) and uric acid (-22%) than those without GH (all p < 0.05). Similarly to high sensitivity C-reactive protein (hsCRP), there were no differences in serum chemokines between children with GH or not (all p > 0.05). Adjusted correlations were significant for RANTES and z-BMI (r = 0.26; p < 0.05) and for MIG with z-BMI (r = -0.26; p < 0.05) and with NEFA (r = 0.27; p < 0.05). CONCLUSION: GH was not associated with higher chemokine levels in prepubertal children with OW/OB. Decreased rather than elevated GFR values were correlated with obesity and worse metabolic profiles. Chemokines levels in children with severe OB suggest a regulation of the immune response. Follow-up studies are needed to address the clinical implications of these findings.


Subject(s)
Chemokines/blood , Child Development , Energy Metabolism , Glomerular Filtration Rate , Kidney Diseases/physiopathology , Kidney/physiopathology , Pediatric Obesity/blood , Age Factors , Biomarkers/blood , Child , Cross-Sectional Studies , Fatty Acids, Nonesterified/blood , Female , Humans , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Male , Pediatric Obesity/complications , Pediatric Obesity/diagnosis , Pediatric Obesity/immunology , Prospective Studies , Risk Factors
12.
Psychoneuroendocrinology ; 116: 104659, 2020 06.
Article in English | MEDLINE | ID: mdl-32240906

ABSTRACT

Childhood obesity constitutes a major global public health challenge. A substantial body of evidence suggests that conditions and states experienced by the embryo/fetus in utero can result in structural and functional changes in cells, tissues, organ systems and homeostatic set points related to obesity. Furthermore, growing evidence suggests that maternal conditions and states experienced prior to conception, such as stress, obesity and metabolic dysfunction, may spill over into pregnancy and influence those key aspects of gestational biology that program offspring obesity risk. In this narrative review, we advance a novel hypothesis and life-span framework to propose that maternal exposure to childhood maltreatment may constitute an important and as-yet-underappreciated risk factor implicated in developmental programming of offspring obesity risk via the long-term psychological, biological and behavioral sequelae of childhood maltreatment exposure. In this context, our framework considers the key role of maternal-placental-fetal endocrine, immune and metabolic pathways and also other processes including epigenetics, oocyte mitochondrial biology, and the maternal and infant microbiomes. Finally, our paper discusses future research directions required to elucidate the nature and mechanisms of the intergenerational transmission of the effects of maternal childhood maltreatment on offspring obesity risk.


Subject(s)
Adult Survivors of Child Abuse , Adverse Childhood Experiences , Fetal Development/physiology , Mother-Child Relations , Pediatric Obesity , Prenatal Exposure Delayed Effects , Female , Humans , Pediatric Obesity/etiology , Pediatric Obesity/immunology , Pediatric Obesity/metabolism , Pediatric Obesity/microbiology , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/microbiology
13.
Diabetes Care ; 43(3): 580-587, 2020 03.
Article in English | MEDLINE | ID: mdl-31937610

ABSTRACT

OBJECTIVE: Sustained excess BMI increases the risk of type 1 diabetes (T1D) in autoantibody-positive relatives without diabetes of patients. We tested whether elevated BMI also accelerates the progression of islet autoimmunity before T1D diagnosis. RESEARCH DESIGN AND METHODS: We studied 706 single autoantibody-positive pediatric TrialNet participants (ages 1.6-18.6 years at baseline). Cumulative excess BMI (ceBMI) was calculated for each participant based on longitudinally accumulated BMI ≥85th age- and sex-adjusted percentile. Recursive partitioning analysis and multivariable modeling defined the age cut point differentiating the risk for progression to multiple positive autoantibodies. RESULTS: At baseline, 175 children (25%) had a BMI ≥85th percentile. ceBMI range was -9.2 to 15.6 kg/m2 (median -1.91), with ceBMI ≥0 kg/m2 corresponding to persistently elevated BMI ≥85th percentile. Younger age increased the progression to multiple autoantibodies, with age cutoff of 9 years defined by recursive partitioning analysis. Although ceBMI was not significantly associated with progression from single to multiple autoantibodies overall, there was an interaction with ceBMI ≥0 kg/m2, age, and HLA (P = 0.009). Among children ≥9 years old without HLA DR3-DQ2 and DR4-DQ8, ceBMI ≥0 kg/m2 increased the rate of progression from single to multiple positive autoantibodies (hazard ratio 7.32, P = 0.004) and conferred a risk similar to that in those with T1D-associated HLA haplotypes. In participants <9 years old, the effect of ceBMI on progression to multiple autoantibodies was not significant regardless of HLA type. CONCLUSIONS: These data support that elevated BMI may exacerbate islet autoimmunity prior to clinical T1D, particularly in children with lower risk based on age and HLA. Interventions to maintain normal BMI may prevent or delay the progression of islet autoimmunity.


Subject(s)
Autoimmunity , Body Mass Index , Diabetes Mellitus, Type 1/etiology , Islets of Langerhans/immunology , Pediatric Obesity/complications , Pediatric Obesity/immunology , Adolescent , Adult , Autoantibodies/blood , Autoantibodies/immunology , Child , Child, Preschool , Cohort Studies , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Progression , Female , Haplotypes , Humans , Infant , Islets of Langerhans/pathology , Male , Middle Aged , Pediatric Obesity/epidemiology , Risk Factors , Young Adult
14.
Pediatr Pulmonol ; 55(3): 809-817, 2020 03.
Article in English | MEDLINE | ID: mdl-31912992

ABSTRACT

Childhood obesity contributes to many diseases, including asthma. There is literature to suggest that asthma developing as a consequence of obesity has a nonallergic or non-T2 phenotype. In this review, obesity-related asthma is utilized as a prototype of non-T2 asthma in children to discuss several nonallergic mechanisms that underlie childhood asthma. Obesity-related asthma is associated with systemic T helper (Th)1 polarization occurring with monocyte activation. These immune responses are mediated by insulin resistance and dyslipidemia, metabolic abnormalities associated with obesity, that are themselves associated with pulmonary function deficits in obese asthmatics. As in other multifactorial diseases, there is both a genetic and an environmental contribution to pediatric obesity-related asthma. In addition to genetic susceptibility, differential DNA methylation is associated with non-T2 immune responses in pediatric obesity-related asthma. Initial investigations into the biology of non-T2 immune responses have identified the upregulation of genes in the CDC42 pathway. CDC42 is a RhoGTPase that plays a key role in Th cell physiology, including preferential naïve Th cell differentiation to Th1 cells, and cytokine production and exocytosis. Although these novel pathways are promising findings to direct targeted therapy development for obesity-related asthma to address the disease burden, there is evidence to suggest that dietary interventions, including diet modification, rather than caloric restriction alone, decrease disease burden. Adoption of a diet rich in micronutrients, including carotenoids and 25-OH cholecalciferol, a vitamin D metabolite, may be beneficial since these are positively correlated with pulmonary function indices, while being protective against metabolic abnormalities associated with the obese asthma phenotype.


Subject(s)
Asthma/etiology , Pediatric Obesity/complications , Animals , Asthma/genetics , Asthma/immunology , Asthma/physiopathology , Child , Epigenomics , Humans , Lung/physiopathology , Metabolic Diseases/immunology , Metabolic Diseases/physiopathology , Pediatric Obesity/genetics , Pediatric Obesity/immunology , Pediatric Obesity/physiopathology
15.
Obes Rev ; 21(2): e12963, 2020 02.
Article in English | MEDLINE | ID: mdl-31663251

ABSTRACT

Obesity epidemic responsible for increase in diabetes, heart diseases, infections and cancer shows no signs of abating. Obesity in children is also on rise, indicating the urgent need of strategies for prevention and intervention that must begin in early life. While originally posited that obesity results from the simple concept of consuming more calories, or genetics, emerging research suggests that the bacteria living in our gut (gut microbiome) and its interactions with immune cells and metabolic organs including adipose tissues (microbiome-immune-metabolic axis) play significant role in obesity development in childhood. Specifically, abnormal changes (dysbiosis) in the gut microbiome, stimulation of inflammatory cytokines, and shifts in the metabolic functions of brown adipose tissue and the browning of white adipose tissue are associated with increased obesity. Many factors from as early as gestation appear to contribute in obesity, such as maternal health, diet, antibiotic use by mother and/or child, and birth and feeding methods. Herein, using evidence from animal and human studies, we discuss how these factors impact microbiome-immune-metabolic axis and cause obesity epidemic in children, and describe the gaps in knowledge that are warranted for future research.


Subject(s)
Gastrointestinal Microbiome/immunology , Pediatric Obesity/immunology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Child , Humans , Pediatric Obesity/metabolism , Pediatric Obesity/microbiology
16.
J Clin Res Pediatr Endocrinol ; 12(1): 63-70, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31434461

ABSTRACT

Objective: Macular damage may be observed in obesity and metabolic syndrome (MetS), a condition which leads to chronic subclinical inflammation and affects most organ systems. To investigate the association between macular variability and anthropometric measurements, metabolic parameters, and inflammatory markers in children and adolescents with MetS. Methods: Two hundred and twenty eyes of 62 obese and 48 healthy children and adolescents were examined. Bilateral macular retinal thickness (MRT) and macular retinal volume (MRV) were measured in all subjects using optical coherence tomography. Associations between mean MRT and mean MRV and age, auxological measurements including body mass index standard deviation scores (BMI-SDS) and waist circumference-SDS (WC-SDS), metabolic parameters and inflammatory parameters including neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio and systemic immune-inflammatory index (SIII) were investigated. Results: No statistically significant difference was observed between the groups in terms of age or sex distribution (p>0.05). Mean MRT (r=-0.326, p=0.007) and MRV (r=-0.303, p=0.007) values in the obese group with MetS decreased as homeostasis model assessment-insulin resistance (HOMA-IR) values increased. SIII values were higher in obese groups, but particularly in obese subject with MetS, compared to the control group (p=0.021). The decrease in mean MRT (r=-0.544, p=0.046) and MRV (r=-0.651, p=0.031) in the obese subjects with MetS was negatively correlated with NLR. Mean MRT and MRV decreased in all obese subjects as SIII increased (p<0.05). Conclusion: This is the first study to show that mean MRT and MRV values decrease as BMI-SDS, WC-SDS and HOMA-IR increase in obese children and adolescents with MetS. NLR and SIII may serve as markers of chronic inflammation in obese children with MetS associated with macular damage.


Subject(s)
Body Mass Index , Inflammation/metabolism , Insulin Resistance , Macula Lutea/pathology , Metabolic Syndrome/metabolism , Pediatric Obesity/metabolism , Retinal Diseases/pathology , Waist Circumference , Adolescent , Biomarkers/metabolism , Child , Cross-Sectional Studies , Female , Humans , Inflammation/blood , Inflammation/immunology , Macula Lutea/diagnostic imaging , Male , Metabolic Syndrome/blood , Metabolic Syndrome/immunology , Pediatric Obesity/blood , Pediatric Obesity/immunology , Retinal Diseases/diagnostic imaging , Tomography, Optical Coherence
17.
J Intellect Disabil Res ; 64(2): 170-179, 2020 02.
Article in English | MEDLINE | ID: mdl-31858639

ABSTRACT

BACKGROUND: The main objective of this study was to describe the inflammatory status of adolescents with Down syndrome (DS) and their relationship with adiposity. METHODS: Ninety-five adolescents with DS (44.2% girls) and a control group of 113 adolescents (47.8% girls), aged between 11 and 18 years old, from the UP & DOWN study were included in this substudy. Serum C-reactive protein, C3 and C4 complement factors, total proteins, interleukin-6, tumour necrosis factor-α, insulin, cortisol, leptin, adiponectin, galactin-3 and visfatin were analysed; homeostatic model assessment index was calculated. In order to evaluate adiposity, we measured the following body fat variables: weight, height, waist circumference and skinfold thicknesses. Birth weight was obtained by questionnaire. In addition, body mass index, waist-to-height ratio (WHtR) and body fat percentage (BF%) were calculated. RESULTS: Down syndrome group showed higher levels of body mass index, WHtR, waist circumference, BF% and lower birth weight than controls (P < 0.001). In the general linear model in the total sample, WHtR was positively associated with C3 and C4 (P < 0.001) as well as with leptin levels (P = 0.015). BF% was positively associated with total proteins (P = 0.093) and leptin levels (P < 0.001). DS was positively associated with total proteins (P < 0.001), C3 (P = 0.047) and C4 (P = 0.019). Despite the higher levels of adiposity found in DS group, no direct association was found between BF% and leptin levels, comparing with the control group. CONCLUSIONS: These findings suggest that abdominal obesity should be controlled in adolescents because of its relationship with acute phase-inflammatory biomarkers but especially in DS adolescents who may show a peculiar metabolic status according to their relationship between adiposity and inflammatory biomarkers.


Subject(s)
Adiposity/physiology , Down Syndrome , Inflammation , Pediatric Obesity , Adiposity/immunology , Adolescent , Biomarkers/blood , Child , Comorbidity , Down Syndrome/epidemiology , Down Syndrome/immunology , Down Syndrome/metabolism , Female , Follow-Up Studies , Humans , Inflammation/epidemiology , Inflammation/immunology , Inflammation/metabolism , Male , Pediatric Obesity/epidemiology , Pediatric Obesity/immunology , Pediatric Obesity/metabolism
18.
Endocr Regul ; 53(2): 71-82, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-31517622

ABSTRACT

OBJECTIVE: The development of obesity and its metabolic complications is associated with dysregulation of various intrinsic mechanisms, which control basic metabolic processes through changes in the expression of numerous regulatory genes. METHODS: The expression level of HLA-DRA, HLA-DRB1, HLA-G, HLA-F, and NFX1 genes as well as miR-190b was measured in the blood of obese adolescents without signs of resistance to insulin and with insulin resistance in comparison with the group of relative healthy control individuals without signs of obesity. RESULTS: It was shown that obesity without signs of insulin resistance is associated with upregulation of the expression level of HLA-DRA and HLA-DRB1 genes, but with down-regulation of HLA-G gene expression in the blood as compared to control group of relative healthy adolescents. At the same time, no significant changes were observed in the expression level of HLA-F and NFX1 genes in the blood of this group of obese adolescents. Development of insulin resistance in obese individuals leads to significant down-regulation of HLA-DRA, HLA-DRB1, HLA-G, and HLA-F gene expressions as well as to up-regulation of NFX1 gene as well as microRNA miR-190b in the blood as compared to obese patients without signs of insulin resistance. CONCLUSIONS: Results of this study provide evidence that obesity affects the expression of the subset of genes related to immune response in the blood and that development of insulin resistance in obese adolescents is associated with strong down-regulation of the expressions of HLA-DRA, HLA-DRB1, HLA-F, and HLA-G genes, which may be contribute to the development of obesity complications. It is possible that transcription factor NFX1 and miR-190b participate in downregulation of HLA-DRA gene expression in the blood of obese adolescents with insulin resistance.


Subject(s)
Immunity, Innate/genetics , Insulin Resistance/physiology , Pediatric Obesity , Adolescent , Blood Cells/metabolism , Case-Control Studies , Female , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-G Antigens/genetics , Histocompatibility Antigens Class I/genetics , Humans , Male , Pediatric Obesity/blood , Pediatric Obesity/genetics , Pediatric Obesity/immunology , Pediatric Obesity/metabolism , Repressor Proteins/genetics
19.
Adv Exp Med Biol ; 1211: 77-87, 2019.
Article in English | MEDLINE | ID: mdl-31456043

ABSTRACT

The prevalence of allergy and obesity is sharply on the rise in children. However, the nature of a mutual relation of the two conditions remains unclear. The aim of the study was to assess the impact of excessive body weight on the immune response in children with allergies. There were 56 children with allergies, aged 4-15 years, included into the study (41 with asthma and 15 with atopic dermatitis). Based on the body mass index, children were divided into two groups: normal weight (body mass index (BMI) <85th percentile) and excessive weight (BMI ≥ 85th percentile). The immunological parameters were evaluated by flow cytometry. We found that children with excessive body weight had a significantly lower percentage of CD4+ lymphocytes and a higher percentage of natural killer T cells (NKT) and CD16/56+ lymphocytes than those with normal weight. In the group with allergy, a significant positive association was noticed between BMI and the percentage of human leukocyte antigen (HLA)-DR-specific CD3. Further analysis was done after dividing the allergy group into the children with normal and excessive weight. There were an adverse association between BMI and the percentage of CD8+ lymphocytes in those with normal weight and a positive one between BMI and the percentage of CD4+ in those with excessive weight. We conclude that excessive body weight plays a major role in mediating the immunological response in children with allergy.


Subject(s)
Asthma/immunology , Body Weight , Dermatitis, Atopic/immunology , Hypersensitivity/immunology , Pediatric Obesity/immunology , Adolescent , Body Mass Index , Child , Child, Preschool , Humans
20.
Int J Obes (Lond) ; 43(4): 713-723, 2019 04.
Article in English | MEDLINE | ID: mdl-30568265

ABSTRACT

BACKGROUND: The gut microbiota has been associated with overweight and obesity in adults, but the evidence in children is limited. Our aim was to study whether composition of the gut microbiota at the age of 3 years is associated with overweight/obesity in children cross-sectionally. METHODS: Children, who participated in a clinical trial of prenatal vitamin-D supplementation (VDAART), underwent standardized height and weight measurements, and collection of stool samples at 3 years of age. 16 S rRNA sequencing (V4 region) of the stool samples were performed with Illumina MiSeq. Associations between microbiota and overweight/obesity (body mass index z-scores >85th percentile) was analyzed using logistic regression. RESULTS: Out of 502 children, 146 (29%) were categorized as overweight/obese. Maternal pre-pregnancy BMI, birth weight and length, formula feeding during the first year, high frequency of fast food consumption, and time watching TV or computer screen at 3 years were the risk factors for overweight/obesity. Of the top 20 most abundant genera, high relative abundance of Parabacteroidetes (Bacteroidetes; Bacteroidales) (aOR(95% CI): 0.69 (0.53, 0.90, p = 0.007) per interquartile increase) and unassigned genus within Peptostreptococcae family were inversely associated with overweight/obesity, whereas high relative abundance of Dorea (Firmicutes;Clostridiales) (1.23 (1.05, 1.43, p = 0.009)) was positively associated. Associations were independent of each other. No associations were found between diversity indices and overweight/obesity. CONCLUSIONS: Our data suggest that some of the differences in gut composition of bacteria between obese and non-obese adults can already be observed in 3-year old children. Longitudinal studies will be needed to determine long-term effects.


Subject(s)
Gastrointestinal Microbiome/physiology , Overweight/physiopathology , Pediatric Obesity/physiopathology , Body Mass Index , Child, Preschool , Cross-Sectional Studies , Female , Follow-Up Studies , Gastrointestinal Microbiome/immunology , Humans , Male , Overweight/immunology , Pediatric Obesity/immunology , Pediatric Obesity/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...