Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Ecol Food Nutr ; 61(6): 728-752, 2022.
Article in English | MEDLINE | ID: mdl-36268551

ABSTRACT

The current mixed-method study uses Food Frequency Questionnaires and 24-hour dietary recalls (n = 41) to assess the food/nutrient intake; and qualitative interviews to identify local perceptions of food among 41 early postpartum women in Belgaum, India. The results show that total energy, protein, and most micronutrient intake were significantly lower than the Recommended Dietary Allowance of India (p < .05 individually); ninety percent of mothers restricted the consumption of some specific fruits, vegetables, and other foods during postpartum due to their perceptions of foods, folk medicines, and health beliefs. Culturally sensitive programs relevant to postpartum diet practices for women should be implemented.


Subject(s)
Diet , Energy Intake , Female , Humans , Seasons , India , Postpartum Period , Vegetables
2.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L116-L128, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34850640

ABSTRACT

Obesity impairs host defense against Klebsiella pneumoniae, but responsible mechanisms are incompletely understood. To determine the impact of diet-induced obesity on pulmonary host defense against K. pneumoniae, we fed 6-wk-old male C57BL/6j mice a normal diet (ND) or high-fat diet (HFD) (13% vs. 60% fat, respectively) for 16 wk. Mice were intratracheally infected with Klebsiella, assayed at 24 or 48 h for bacterial colony-forming units, lung cytokines, and leukocytes from alveolar spaces, lung parenchyma, and gonadal adipose tissue were assessed using flow cytometry. Neutrophils from uninfected mice were cultured with and without 2-deoxy-d-glucose (2-DG) and assessed for phagocytosis, killing, reactive oxygen intermediates (ROI), transport of 2-DG, and glucose transporter (GLUT1-4) transcripts, and protein expression of GLUT1 and GLUT3. HFD mice had higher lung and splenic bacterial burdens. In HFD mice, baseline lung homogenate concentrations of IL-1ß, IL-6, IL-17, IFN-γ, CXCL2, and TNF-α were reduced relative to ND mice, but following infection were greater for IL-6, CCL2, CXCL2, and IL-1ß (24 h only). Despite equivalent lung homogenate leukocytes, HFD mice had fewer intraalveolar neutrophils. HFD neutrophils exhibited decreased Klebsiella phagocytosis and killing and reduced ROI to heat-killed Klebsiella in vitro. 2-DG transport was lower in HFD neutrophils, with reduced GLUT1 and GLUT3 transcripts and protein (GLUT3 only). Blocking glycolysis with 2-DG impaired bacterial killing and ROI production in neutrophils from mice fed ND but not HFD. Diet-induced obesity impairs pulmonary Klebsiella clearance and augments blood dissemination by reducing neutrophil killing and ROI due to impaired glucose transport.


Subject(s)
Diet , Glucose/metabolism , Host-Pathogen Interactions , Klebsiella Infections/microbiology , Klebsiella pneumoniae/physiology , Neutrophils/metabolism , Obesity/microbiology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Bacterial Load/drug effects , Biological Transport/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Bone Marrow/pathology , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Deoxyglucose/pharmacology , Diet, High-Fat , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glycolysis/drug effects , Host-Pathogen Interactions/drug effects , Klebsiella Infections/blood , Klebsiella Infections/complications , Klebsiella pneumoniae/drug effects , Leukocyte Count , Lung/microbiology , Lung/pathology , Male , Mice, Inbred C57BL , Neutrophils/drug effects , Obesity/blood , Obesity/complications , Phagocytosis/drug effects , Pneumonia/microbiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/microbiology
3.
J Am Heart Assoc ; 10(7): e019173, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33779242

ABSTRACT

Background The menopausal transition is characterized by increased cardiovascular risk, weight gain, and increased adiposity for many women. The adipose-derived secretory proteins adiponectin and leptin are associated with insulin resistance, metabolic syndrome, and cardiovascular disease but their role in subclinical atherosclerotic disease is unclear. This cross-sectional study evaluated the associations of adiponectin and leptin with carotid artery intima-media thickness, adventitial diameter, presence of carotid plaques, and brachial-ankle pulse wave velocity (baPWV) in women aged 54 to 65 years. Methods and Results Participants were 1399 women from SWAN (Study of Women's Health Across the Nation), a community-based study of women transitioning through menopause. Carotid ultrasound and baPWV measures were obtained at SWAN follow-up visits 12 or 13, when 97% of participants were post-menopausal. Adipokines were assayed from serum specimens obtained concurrently at these visits. Linear and logistic regression models were used to evaluate adiponectin or leptin, both log-transformed attributable to skewness, in relationship to carotid artery intima-media thickness, adventitial diameter, baPWV, and presence of carotid plaque. Covariates included age, race, study site, smoking, alcohol use, obesity, cardiovascular disease risk factors, and menopausal status. Lower levels of adiponectin were related to greater carotid artery intima-media thickness, wider adventitial diameter, and faster baPWV; associations were attenuated after adjusting for cardiovascular disease risk factors. Higher levels of leptin were associated with greater carotid artery intima-media thickness and wider adventitial diameter in minimally and fully adjusted models, and contrary to expectation, with slower baPWV, particularly among women with diabetes mellitus or obesity. Conclusions Adiponectin and leptin are 2 important inflammatory pathways that may contribute to adverse subclinical cardiovascular disease risk profiles in women at midlife.


Subject(s)
Adipokines/blood , Cardiovascular Diseases/blood , Ethnicity , Postmenopause/blood , Women's Health , Adult , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/ethnology , Carotid Intima-Media Thickness , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , Incidence , Middle Aged , Risk Factors , Ultrasonography , United States/epidemiology
4.
Life Sci Alliance ; 3(11)2020 11.
Article in English | MEDLINE | ID: mdl-32820026

ABSTRACT

Alveolar macrophages (AMs) are resident immune cells of the lung that are critical for host defense. AMs are capable of proliferative renewal, yet their numbers are known to decrease with aging and increase with cigarette smoking. The mechanism by which AM proliferation is physiologically restrained, and whether dysregulation of this brake contributes to altered AM numbers in pathologic circumstances, however, remains unknown. Mice of advanced age exhibited diminished basal AM numbers and contained elevated PGE2 levels in their bronchoalveolar lavage fluid (BALF) as compared with young mice. Exogenous PGE2 inhibited AM proliferation in an E prostanoid receptor 2 (EP2)-cyclic AMP-dependent manner. Furthermore, EP2 knockout (EP2 KO) mice exhibited elevated basal AM numbers, and their AMs resisted the ability of PGE2 and aged BALF to inhibit proliferation. In contrast, increased numbers of AMs in mice exposed to cigarette smoking were associated with reduced PGE2 levels in BALF and were further exaggerated in EP2 KO mice. Collectively, our findings demonstrate that PGE2 functions as a tunable brake on AM numbers under physiologic and pathophysiological conditions.


Subject(s)
Macrophages, Alveolar/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Aging/physiology , Animals , Bronchoalveolar Lavage Fluid/immunology , Dinoprostone/metabolism , Dinoprostone/physiology , Female , Lung/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/physiology , Smoking/adverse effects
5.
Cells ; 9(7)2020 06 30.
Article in English | MEDLINE | ID: mdl-32630102

ABSTRACT

Extracellular vesicles (EVs) contain a diverse array of molecular cargoes that alter cellular phenotype and function following internalization by recipient cells. In the lung, alveolar macrophages (AMs) secrete EVs containing suppressor of cytokine signaling 3 (SOCS3), a cytosolic protein that promotes homeostasis via vesicular transfer to neighboring alveolar epithelial cells. Although changes in the secretion of EV molecules-including but not limited to SOCS3-have been described in response to microenvironmental stimuli, the cellular and molecular machinery that control alterations in vesicular cargo packaging remain poorly understood. Furthermore, the use of quantitative methods to assess the sorting of cytosolic cargo molecules into EVs is lacking. Here, we utilized cigarette smoke extract (CSE) exposure of AMs as an in vitro model of oxidative stress to address these gaps in knowledge. We demonstrate that the accumulation of reactive oxygen species (ROS) in AMs was sufficient to augment vesicular SOCS3 release in this model. Using nanoparticle tracking analysis (NTA) in tandem with a new carboxyfluorescein succinimidyl ester (CFSE)-based intracellular protein packaging assay, we show that the stimulatory effects of CSE were at least in part attributable to elevated amounts of SOCS3 packaged per EV secreted by AMs. Furthermore, the use of a 20S proteasome activity assay alongside treatment of AMs with conventional proteasome inhibitors strongly suggest that ROS stimulated SOCS3 release via inactivation of the proteasome. These data demonstrate that tuning of AM proteasome function by microenvironmental oxidants is a critical determinant of the packaging and secretion of cytosolic SOCS3 protein within EVs.


Subject(s)
Extracellular Vesicles/metabolism , Macrophages, Alveolar/metabolism , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Cells, Cultured , Female , Macrophages, Alveolar/drug effects , Oxidants/toxicity , Oxidative Stress , Rats , Rats, Wistar , Secretory Pathway , Tobacco Smoke Pollution/adverse effects
6.
Arthritis Care Res (Hoboken) ; 72(7): 874-881, 2020 07.
Article in English | MEDLINE | ID: mdl-31074595

ABSTRACT

OBJECTIVE: To examine associations between dietary intake of omega-3 (n-3; generally antiinflammatory) and omega-6 (n-6; generally proinflammatory) fatty acids and patient-reported outcomes in systemic lupus erythematosus (SLE). METHODS: This study was based on the population-based Michigan Lupus Epidemiology and Surveillance cohort. Estimates of n-3 and n-6 intake were derived from Diet History Questionnaire II items (past year with portion size version). Patient-reported outcomes included self-reported lupus activity (Systemic Lupus Activity Questionnaire [SLAQ]). Multivariable regression, adjusted for age, sex, race, and body mass index, was used to assess associations between absolute intake of n-3 and n-6, as well as the n-6:n-3 ratio, and patient-reported outcomes. RESULTS: Among 456 SLE cases, 425 (93.2%) were female, 207 (45.4%) were African American, and the mean ± SD age was 52.9 ± 12.3 years. Controlling for potential confounders, the average SLAQ score was significantly higher by 0.3 points (95% confidence interval [95% CI] 0.1, 0.6; P = 0.013) with each unit increase of the n-6:n-3 ratio. Both lupus activity and Patient-Reported Outcomes Measurement Information System (PROMIS) sleep disturbance scores were lower with each 1-gram/1,000 kcal increase of n-3 fatty acids (SLAQ regression coefficient ß = -0.8 [95% CI -1.6, 0.0]; P = 0.055; PROMIS sleep ß = -1.1 [95% CI -2.0, -0.2]; P = 0.017). Higher n-3 intakes were nonsignificantly associated with lower levels of depressive symptoms and comorbid fibromyalgia, and with higher quality of life, whereas results for the n6:n3 ratio trended in the opposite direction. CONCLUSION: This population-based study suggests that higher dietary intake of n-3 fatty acids and lower n-6:n-3 ratios are favorably associated with patient-reported outcomes in SLE, particularly self-reported lupus activity and sleep quality.


Subject(s)
Diet , Fatty Acids, Omega-3 , Fatty Acids, Omega-6 , Lupus Erythematosus, Systemic , Adult , Aged , Cohort Studies , Diet Surveys , Female , Humans , Male , Michigan , Middle Aged , Patient Reported Outcome Measures , Quality of Life
7.
Article in English | MEDLINE | ID: mdl-30915034

ABSTRACT

During the last 40 years, there has been a world-wide increase in both the prevalence of obesity and an increase in the number of persons over the age of 60 due to a decline in deaths from infectious disease and the nutrition transition in low and middle income nations. While the increase in the elderly population indicates improvements in global public health, this population may experience a diminished quality of life due to the negative impacts of obesity on age-associated inflammation. Aging alters adipose tissue composition and function resulting in insulin resistance and ectopic lipid storage. A reduction in brown adipose tissue activity, declining sex hormones levels, and abdominal adipose tissue expansion occur with advancing years through the redistribution of lipids from the subcutaneous to the visceral fat compartment. These changes in adipose tissue function and distribution influence the secretion of adipose tissue derived hormones, or adipokines, that promote a chronic state of low-grade systemic inflammation. Ultimately, obesity accelerates aging by enhancing inflammation and increasing the risk of age-associated diseases. The focus of this review is the impact of aging on adipose tissue distribution and function and how these effects influence the elaboration of pro and anti-inflammatory adipokines.

8.
Psychoneuroendocrinology ; 97: 20-27, 2018 11.
Article in English | MEDLINE | ID: mdl-30005278

ABSTRACT

Small clinical studies suggest depression is associated with alterations in adiponectin and leptin, adipocyte-derived secretory proteins involved in metabolic regulation; however, longitudinal data on these association are lacking. This study examined cross-sectional and longitudinal associations of depressive symptoms and major depressive disorder (MDD) with adiponectin and leptin in healthy middle-aged women (mean (SD) age, 45.6 (2.5) years). Cross-sectional analyses included 575 women with baseline adipokine data; longitudinal analyses included 262 women with 2-4 adipokine measurements over 5 years. The 20-item Center for Epidemiologic Studies Depression scale (CES-D) was used to assess depressive symptoms; history of MDD was determined by the Structured Clinical Interview for DSM-IV. Adipokines were assayed from stored serum specimens; values were log-transformed for analyses. Linear and repeated measure random effects regression models evaluated associations of baseline CES-D scores with baseline adipokine concentrations and changes over time, respectively. Secondary analyses evaluated the relation of MDD history with adipokine concentrations. Mean (SD) baseline concentrations of adiponectin and leptin were 9.90 (4.92) µg/mL and 27.02 (20.06) ng/mL; both increased over time (p < .0001). CES-D scores were associated with lower adiponectin at baseline (per 1-SD: estimate=-0.04, SE=.02, p=.03) and over time (per 1-SD: estimate=-0.055, SE = .024, p=.02). Associations were unchanged in risk factor-adjusted models. Women with elevated CES-D scores (≥16) had 6.9% (95% CI: -1.1%, 14.3%; p = .089) lower median adiponectin at baseline and 11.5% (95% CI: 1.5%, 20.4%, p = .025) lower median adiponectin over time in adjusted models, compared to women with CES-D<16. Rate of change in adipokines did not vary by baseline depressive symptoms or MDD history. Depressive symptoms and MDD history were unrelated to leptin. In women at midlife, depressive symptoms are associated with lower adiponectin, a critical anti-inflammatory biomarker involved in metabolic and cardiovascular conditions.


Subject(s)
Adipokines/analysis , Adiponectin/analysis , Depression/metabolism , Adipokines/metabolism , Adiponectin/metabolism , Adult , Cross-Sectional Studies , Depressive Disorder, Major/metabolism , Female , Humans , Leptin/analysis , Longitudinal Studies , Middle Aged , Psychological Tests , Women's Health
9.
Am J Physiol Lung Cell Mol Physiol ; 315(1): L78-L86, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29565180

ABSTRACT

Leptin is a pleiotropic hormone produced by white adipose tissue that regulates appetite and many physiological functions, including the immune response to infection. Genetic leptin deficiency in humans and mice impairs host defenses against respiratory tract infections. Since leptin deficiency is associated with obesity and other metabolic abnormalities, we generated mice that lack the leptin receptor (LepRb) in cells of the myeloid linage (LysM-LepRb-KO) to evaluate its impact in lean metabolically normal mice in a murine model of pneumococcal pneumonia. We observed higher lung and spleen bacterial burdens in LysM-LepRb-KO mice following an intratracheal challenge with Streptococcus pneumoniae. Although numbers of leukocytes recovered from bronchoalveolar lavage fluid did not differ between groups, we did observe higher levels of pulmonary IL-13 and TNFα in LysM-LepRb-KO mice 48 h post infection. Phagocytosis and killing of ingested S. pneumoniae were also impaired in alveolar macrophages (AMs) from LysM-LepRb-KO mice in vitro and were associated with reduced LTB4 and enhanced PGE2 synthesis in vitro. Pretreatment of AMs with LTB4 and the cyclooxygenase inhibitor, indomethacin, restored phagocytosis but not bacterial killing in vitro. These results confirm our previous observations in leptin-deficient ( ob/ob) and fasted mice and demonstrate that decreased leptin action, as opposed to metabolic irregularities associated with obesity or starvation, is responsible for the defective host defense against pneumococcal pneumonia. They also provide novel targets for therapeutic intervention in humans with bacterial pneumonia.


Subject(s)
Lung/immunology , Macrophages/immunology , Phagocytosis , Pneumonia, Pneumococcal/immunology , Receptors, Leptin/immunology , Streptococcus pneumoniae/immunology , Animals , Interleukin-13/genetics , Interleukin-13/immunology , Lung/microbiology , Lung/pathology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/pathology , Receptors, Leptin/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Chem Res Toxicol ; 30(7): 1384-1395, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28574698

ABSTRACT

From 1999-2014, obesity prevalence increased among adults and youth. Obese individuals may be uniquely susceptible to the proinflammatory effects of ozone because obese humans and animals have been shown to experience a greater decline in lung function than normal-weight subjects. Obesity is independently associated with limitations in lung mechanics with increased ozone dose. However, few epidemiologic studies have examined the interaction between excess weight and ozone exposure among adults. Using PubMed keyword searches and reference lists, we reviewed epidemiologic evidence to identify potential response-modifying factors and determine if obese or overweight adults are at increased risk of ozone-related health effects. We initially identified 170 studies, of which seven studies met the criteria of examining the interaction of excess weight and ozone exposure on cardiopulmonary outcomes in adults, including four short-term ozone exposure studies in controlled laboratory settings and three community epidemiologic studies. In the studies identified, obesity was associated with decreased lung function and increased inflammatory mediators. Results were inconclusive about the effect modification when data were stratified by sex. Obese and overweight populations should be considered as candidate at-risk groups for epidemiologic studies of cardiopulmonary health related to air pollution exposures. Air pollution is a modifiable risk factor that may decrease lung function among obese individuals with implications for environmental and occupational health policy.


Subject(s)
Air Pollutants/adverse effects , Cardiovascular Diseases/pathology , Environmental Exposure/adverse effects , Lung/pathology , Obesity/pathology , Ozone/adverse effects , Animals , Humans , Ozone/administration & dosage
11.
Immunotargets Ther ; 5: 47-56, 2016.
Article in English | MEDLINE | ID: mdl-27529061

ABSTRACT

Adipose tissue has traditionally been defined as connective tissue that stores excess calories in the form of triacylglycerol. However, the physiologic functions attributed to adipose tissue are expanding, and it is now well established that adipose tissue is an endocrine gland. Among the endocrine factors elaborated by adipose tissue are the adipokines; hormones, similar in structure to cytokines, produced by adipose tissue in response to changes in adipocyte triacylglycerol storage and local and systemic inflammation. They inform the host regarding long-term energy storage and have a profound influence on reproductive function, blood pressure regulation, energy homeostasis, the immune response, and many other physiologic processes. The adipokines possess pro- and anti-inflammatory properties and play a critical role in integrating systemic metabolism with immune function. In calorie restriction and starvation, proinflammatory adipokines decline and anti-inflammatory adipokines increase, which informs the host of energy deficits and contributes to the suppression of immune function. In individuals with normal metabolic status, there is a balance of pro- and anti-inflammatory adipokines. This balance shifts to favor proinflammatory mediators as adipose tissue expands during the development of obesity. As a consequence, the proinflammatory status of adipose tissue contributes to a chronic low-grade state of inflammation and metabolic disorders associated with obesity. These disturbances are associated with an increased risk of metabolic disease, type 2 diabetes, cardiovascular disease, and many other pathological conditions. This review focuses on the impact of energy homeostasis on the adipokines in immune function.

12.
J Immunol ; 196(12): 5112-20, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183597

ABSTRACT

Preservation of gas exchange mandates that the pulmonary alveolar surface restrain unnecessarily harmful inflammatory responses to the many challenges to which it is exposed. These responses reflect the cross-talk between alveolar epithelial cells (AECs) and resident alveolar macrophages (AMs). We recently determined that AMs can secrete suppressor of cytokine signaling (SOCS) proteins within microparticles. Uptake of these SOCS-containing vesicles by epithelial cells inhibits cytokine-induced STAT activation. However, the ability of epithelial cells to direct AM release of SOCS-containing vesicles in response to inflammatory insults has not been studied. In this study, we report that SOCS3 protein was elevated in bronchoalveolar lavage fluid of both virus- and bacteria-infected mice, as well as in an in vivo LPS model of acute inflammation. In vitro studies revealed that AEC-conditioned medium (AEC-CM) enhanced AM SOCS3 secretion above basal levels. Increased amounts of PGE2 were present in AEC-CM after LPS challenge, and both pharmacologic inhibition of PGE2 synthesis in AECs and neutralization of PGE2 in AEC-CM implicated this prostanoid as the major AEC-derived factor mediating enhanced AM SOCS3 secretion. Moreover, pharmacologic blockade of PGE2 synthesis or genetic deletion of a PGE2 synthase similarly attenuated the increase in bronchoalveolar lavage fluid SOCS3 noted in lungs of mice challenged with LPS in vivo. These results demonstrate a novel tunable form of cross-talk in which AECs use PGE2 as a signal to request SOCS3 from AMs to dampen their endogenous inflammatory responses during infection.


Subject(s)
Alveolar Epithelial Cells/metabolism , Bronchoalveolar Lavage Fluid/immunology , Dinoprostone/metabolism , Immunity, Innate , Macrophages, Alveolar/immunology , Suppressor of Cytokine Signaling 3 Protein/metabolism , Alveolar Epithelial Cells/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Cell Line, Tumor , Cells, Cultured , Culture Media , Inflammation , Lipopolysaccharides/immunology , Macrophages, Alveolar/metabolism , Mice , Prostaglandin-E Synthases/deficiency , Prostaglandin-E Synthases/genetics , Rats , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/immunology
13.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1111-20, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27059285

ABSTRACT

The production of prostaglandin E2 (PGE2) increases dramatically during pneumococcal pneumonia, and this lipid mediator impairs alveolar macrophage (AM)-mediated innate immune responses. Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme involved in the synthesis of PGE2, and its expression is enhanced during bacterial infections. Genetic deletion of mPGES-1 in mice results in diminished PGE2 production and elevated levels of other prostaglandins after infection. Since PGE2 plays an important immunoregulatory role during bacterial pneumonia we assessed the impact of mPGES-1 deletion in the host defense against pneumococcal pneumonia in vivo and in AMs in vitro. Wild-type (WT) and mPGES-1 knockout (KO) mice were challenged with Streptococcus pneumoniae via the intratracheal route. Compared with WT animals, we observed reduced survival and increased lung and spleen bacterial burdens in mPGES-1 KO mice 24 and 48 h after S. pneumoniae infection. While we found modest differences between WT and mPGES-1 KO mice in pulmonary cytokines, AMs from mPGES-1 KO mice exhibited defective killing of ingested bacteria in vitro that was associated with diminished inducible nitric oxide synthase expression and reduced nitric oxide (NO) synthesis. Treatment of AMs from mPGES-1 KO mice with an NO donor restored bacterial killing in vitro. These results suggest that mPGES-1 plays a critical role in bacterial pneumonia and that genetic ablation of this enzyme results in diminished pulmonary host defense in vivo and in vitro. These results suggest that specific inhibition of PGE2 synthesis by targeting mPGES-1 may weaken host defense against bacterial infections.


Subject(s)
Cyclooxygenase 1/genetics , Membrane Proteins/genetics , Pneumonia, Pneumococcal/enzymology , Streptococcus pneumoniae/immunology , Animals , Cytokines/biosynthesis , Cytokines/blood , Dinoprostone/biosynthesis , Female , Immunity, Innate , Lung/enzymology , Lung/immunology , Lung/microbiology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/immunology , Mice, Inbred C57BL , Mice, Knockout , Microsomes/enzymology , Nitric Oxide/biosynthesis , Pneumonia, Pneumococcal/immunology
14.
J Gerontol A Biol Sci Med Sci ; 71(4): 508-14, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26302979

ABSTRACT

BACKGROUND: Excess fat mass is a greater contributor to functional limitations than is reduced lean mass or the presence of obesity-related conditions. The impact of fat mass on physical functioning may be due to adipokines, adipose-derived proteins that have pro- or anti-inflammatory properties. METHODS: Serum samples from 1996 to 2003 that were assayed for leptin, adiponectin, and resistin were provided by 511 participants from the Michigan site of the Study of Women's Health Across the Nation. Physical functioning performance was assessed annually during study visits from 1996 to 2003. RESULTS: Among this population of Black and White women (mean baseline age = 45.6 years, SD = 2.7 years), all of whom were premenopausal at baseline, higher baseline leptin concentrations predicted longer stair climb, sit-to-rise, and 2-pound lift times and shorter forward reach distance (all p < .01). This relationship persisted after adjustment for age, BMI, percent skeletal muscle mass, race/ethnicity, economic strain, bodily pain, diabetes, knee osteoarthritis, and C-reactive protein. Baseline total adiponectin concentrations did not predict any mobility measures but did predict quadriceps strength; a 1 µg/mL higher adiponectin concentration was associated with 0.64 Nm lower quadriceps strength (p = .02). Resistin was not associated with any of the physical functioning performance measures. Change in the adipokines was not associated with physical functioning. CONCLUSION: In this population of middle-aged women, higher baseline leptin concentrations predicted poorer mobility-based functioning, whereas higher adiponectin concentrations predicted reduced quadriceps strength. These findings suggest that the relationship between the adipokines and physical functioning performance is independent of other known correlates of poor functioning.


Subject(s)
Adiponectin/blood , Leptin/blood , Physical Fitness/physiology , Female , Humans , Longitudinal Studies , Michigan , Middle Aged , Muscle Strength/physiology , Predictive Value of Tests , Resistin/blood , United States
15.
J Immunol ; 195(1): 174-84, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25987742

ABSTRACT

Inhaled corticosteroids (ICS) increase community-acquired pneumonia (CAP) incidence in patients with chronic obstructive pulmonary disease (COPD) by unknown mechanisms. Apoptosis is increased in the lungs of COPD patients. Uptake of apoptotic cells (ACs) ("efferocytosis") by alveolar macrophages (AMøs) reduces their ability to combat microbes, including Streptococcus pneumoniae, the most common cause of CAP in COPD patients. Having shown that ICS significantly increase AMø efferocytosis, we hypothesized that this process, termed glucocorticoid-augmented efferocytosis, might explain the association of CAP with ICS therapy in COPD. To test this hypothesis, we studied the effects of fluticasone, AC, or both on AMøs of C57BL/6 mice in vitro and in an established model of pneumococcal pneumonia. Fluticasone plus AC significantly reduced TLR4-stimulated AMø IL-12 production, relative to either treatment alone, and decreased TNF-α, CCL3, CCL5, and keratinocyte-derived chemoattractant/CXCL1, relative to AC. Mice treated with fluticasone plus AC before infection with viable pneumococci developed significantly more lung CFUs at 48 h. However, none of the pretreatments altered inflammatory cell recruitment to the lungs at 48 h postinfection, and fluticasone plus AC less markedly reduced in vitro mediator production to heat-killed pneumococci. Fluticasone plus AC significantly reduced in vitro AMø killing of pneumococci, relative to other conditions, in part by delaying phagolysosome acidification without affecting production of reactive oxygen or nitrogen species. These results support glucocorticoid-augmented efferocytosis as a potential explanation for the epidemiological association of ICS therapy of COPD patients with increased risk for CAP, and establish murine experimental models to dissect underlying molecular mechanisms.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Androstadienes/adverse effects , Lung/immunology , Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Animals , Apoptosis , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Colony Count, Microbial , Disease Models, Animal , Fluticasone , Gene Expression Regulation , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Phagocytosis , Pneumonia, Pneumococcal/chemically induced , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/microbiology , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , Streptococcus pneumoniae/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
16.
J Immunol ; 194(10): 4846-59, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25862817

ABSTRACT

Obese individuals are at greater risk for hospitalization and death from infection with the 2009 pandemic H1N1 influenza virus (pH1N1). In this study, diet-induced and genetic-induced obese mouse models were used to uncover potential mechanisms by which obesity increases pH1N1 severity. High-fat diet-induced and genetic-induced obese mice exhibited greater pH1N1 mortality, lung inflammatory responses, and excess lung damage despite similar levels of viral burden compared with lean control mice. Furthermore, obese mice had fewer bronchoalveolar macrophages and regulatory T cells during infection. Obesity is inherently a metabolic disease, and metabolic profiling has found widespread usage in metabolic and infectious disease models for identifying biomarkers and enhancing understanding of complex mechanisms of disease. To further characterize the consequences of obesity on pH1N1 infection responses, we performed global liquid chromatography-mass spectrometry metabolic profiling of lung tissue and urine. A number of metabolites were perturbed by obesity both prior to and during infection. Uncovered metabolic signatures were used to identify changes in metabolic pathways that were differentially altered in the lungs of obese mice such as fatty acid, phospholipid, and nucleotide metabolism. Taken together, obesity induces distinct alterations in the lung metabolome, perhaps contributing to aberrant pH1N1 immune responses.


Subject(s)
Lung/metabolism , Lung/pathology , Obesity/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Animals , Chromatography, Liquid , Disease Models, Animal , Flow Cytometry , Influenza A Virus, H1N1 Subtype , Male , Mass Spectrometry , Metabolome , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications
17.
J Exp Med ; 212(5): 729-42, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25847945

ABSTRACT

JAK-STAT signaling mediates the actions of numerous cytokines and growth factors, and its endogenous brake is the family of SOCS proteins. Consistent with their intracellular roles, SOCS proteins have never been identified in the extracellular space. Here we report that alveolar macrophages can secrete SOCS1 and -3 in exosomes and microparticles, respectively, for uptake by alveolar epithelial cells and subsequent inhibition of STAT activation. Secretion is tunable and occurs both in vitro and in vivo. SOCS secretion into lung lining fluid was diminished by cigarette smoking in humans and mice. Secretion and transcellular delivery of vesicular SOCS proteins thus represent a new model for the control of inflammatory signaling, which is subject to dysregulation during states of inflammation.


Subject(s)
Cell-Derived Microparticles/immunology , Epithelial Cells/immunology , Macrophages/immunology , Pulmonary Alveoli/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Animals , Cell Line, Transformed , Cell-Derived Microparticles/pathology , Epithelial Cells/pathology , Female , Humans , Inflammation/immunology , Inflammation/pathology , Janus Kinases/immunology , Male , Mice , Pulmonary Alveoli/pathology , Rats , Rats, Wistar , STAT Transcription Factors/immunology
18.
J Allergy (Cairo) ; 2015: 157919, 2015.
Article in English | MEDLINE | ID: mdl-26770217

ABSTRACT

Background. The prevalence of obesity has increased dramatically over the last decades, and its association with asthma is being increasingly recognized. Aims. Our hypothesis is that increased leptin and decreased adiponectin levels in obese subjects play a direct role in regulating inflammation in asthmatics. We wanted to examine the hypothesis that cysteinyl leukotrienes (cys-LT), inflammatory mediators that are regulated by adipokines, are involved in the pathogenesis of asthma. Methods. We studied a population of asthmatics and nonasthmatics, who in turn were divided into obese and nonobese categories. We examined leptin and its ratio to adiponectin, in asthmatics and nonasthmatics, with and without obesity. In addition, we measured cys-LT levels in exhaled breath condensate (EBC) and in peripheral blood monocytes (PBM) in these groups. Results. Leptin levels were increased in obese asthmatics compared to obese nonasthmatics. The leptin/adiponectin (L/A) ratio was higher in obese asthmatics compared to obese nonasthmatics. EBC cys-LT levels were elevated in asthmatics compared to nonasthmatics. Discussion. Proinflammatory adipokines, released from adipose tissue, may promote an asthma phenotype through enhanced cys-LT production that may result in more prevalent and difficult to control airway disease.

19.
PLoS One ; 9(9): e106420, 2014.
Article in English | MEDLINE | ID: mdl-25203099

ABSTRACT

In the US and globally, dramatic increases in the prevalence of adult and childhood obesity have been reported during the last 30 years. In addition to cardiovascular disease, type II diabetes, and liver disease, obesity has recently been recognized as an important risk factor for influenza pneumonia. During the influenza pandemic of 2009, obese individuals experienced a greater severity of illness from the H1N1 virus. In addition, obese mice have also been shown to exhibit increased lethality and aberrant pulmonary inflammatory responses following influenza infection. In contrast to influenza, the impact of obesity on bacterial pneumonia in human patients is controversial. In this report, we compared the responses of lean WT and obese CPE(fat/fat) mice following an intratracheal infection with Streptococcus pneumoniae, the leading cause of community-acquired pneumonia. At 16 weeks of age, CPE(fat/fat) mice develop severe obesity, hyperglycemia, elevated serum triglycerides and leptin, and increased blood neutrophil counts. There were no differences between lean WT and obese CPE(fat/fat) mice in survival or lung and spleen bacterial burdens following intratracheal infection with S. pneumoniae. Besides a modest increase in TNF-α levels and increased peripheral blood neutrophil counts in CPE(fat/fat) mice, there were not differences in lung or serum cytokines after infection. These results suggest that obesity, accompanied by hyperglycemia and modestly elevated triglycerides, at least in the case of CPE(fat/fat) mice, does not impair innate immunity against pneumococcal pneumonia.


Subject(s)
Immunity, Innate , Obesity/immunology , Obesity/microbiology , Streptococcus pneumoniae/physiology , Animals , Body Weight , Carboxypeptidases/metabolism , Female , Humans , Mice , Mice, Obese , Obesity/metabolism
20.
J Psychiatr Res ; 57: 58-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24953860

ABSTRACT

Polyunsaturated fatty acids (PUFA) profiles associate with risk for mood disorders. This poses the hypothesis of metabolic differences between patients and unaffected healthy controls that relate to the primary illness or are secondary to medication use or dietary intake. However, dietary manipulation or supplementation studies show equivocal results improving mental health outcomes. This study investigates dietary patterns and metabolic profiles relevant to PUFA metabolism, in bipolar I individuals compared to non-psychiatric controls. We collected seven-day diet records and performed metabolomic analysis of fasted plasma collected immediately after diet recording. Regression analyses adjusted for age, gender and energy intake found that bipolar individuals had significantly lower intake of selenium and PUFAs, including eicosapentaenoic acid (EPA) (n-3), docosahexaenoic acid (DHA) (n-3), arachidonic acid (AA) (n-6) and docosapentaenoic acid (DPA) (n-3/n-6 mix); and significantly increased intake of the saturated fats, eicosanoic and docosanoic acid. Regression analysis of metabolomic data derived from plasma samples, correcting for age, gender, BMI, psychiatric medication use and dietary PUFA intake, revealed that bipolar individuals had reduced 13S-HpODE, a major peroxidation product of the n-6, linoleic acid (LA), reduced eicosadienoic acid (EDA), an elongation product of LA; reduced prostaglandins G2, F2 alpha and E1, synthesized from n-6 PUFA; and reduced EPA. These observations remained significant or near significant after Bonferroni correction and are consistent with metabolic variances between bipolar and control individuals with regard to PUFA metabolism. These findings suggest that specific dietary interventions aimed towards correcting these metabolic disparities may impact health outcomes for individuals with bipolar disorder.


Subject(s)
Bipolar Disorder/metabolism , Dietary Fats/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/blood , Adult , Arachidonic Acid/administration & dosage , Arachidonic Acid/blood , Bipolar Disorder/blood , Diet Surveys/statistics & numerical data , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/blood , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/blood , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/blood , Female , Humans , Linoleic Acid/administration & dosage , Linoleic Acid/blood , Male , Middle Aged , Regression Analysis , Risk Factors , Selenium/administration & dosage , Selenium/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...