Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 158(1): 25-40, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995976

ABSTRACT

Obesity and diabetes affect more than half a billion individuals worldwide. Interestingly, the two conditions do not always coincide and the molecular determinants of "healthy" versus "unhealthy" obesity remain ill-defined. Chronic metabolic inflammation (metaflammation) is believed to be pivotal. Here, we tested a hypothesized anti-inflammatory role for heme oxygenase-1 (HO-1) in the development of metabolic disease. Surprisingly, in matched biopsies from "healthy" versus insulin-resistant obese subjects we find HO-1 to be among the strongest positive predictors of metabolic disease in humans. We find that hepatocyte and macrophage conditional HO-1 deletion in mice evokes resistance to diet-induced insulin resistance and inflammation, dramatically reducing secondary disease such as steatosis and liver toxicity. Intriguingly, cellular assays show that HO-1 defines prestimulation thresholds for inflammatory skewing and NF-κB amplification in macrophages and for insulin signaling in hepatocytes. These findings identify HO-1 inhibition as a potential therapeutic strategy for metabolic disease.


Subject(s)
Heme Oxygenase-1/metabolism , Insulin Resistance , Membrane Proteins/metabolism , Obesity/complications , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Liver/metabolism , Macrophages/metabolism , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Mice , Mice, Knockout , Obesity/physiopathology , Reactive Oxygen Species/metabolism
2.
Mol Cell ; 75(3): 644-660.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398325

ABSTRACT

Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.


Subject(s)
Cell Communication/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Sequence Analysis, RNA , Animals , Cellular Reprogramming/genetics , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligands , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/genetics , Single-Cell Analysis
3.
Article in English | MEDLINE | ID: mdl-39189801

ABSTRACT

Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and employs angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine if prior RV-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 x 105 TCID50 units HCoV-NL63. Wild-type mice were infected with 1 x 106 PFU RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 non-structural protein 3 (nsp3), and host gene expression by next generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection four days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked three days after infection and inflammation persisted six days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins and pro-inflammatory cytokines. Infection with RV-A1B four days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared to sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.

4.
Cell ; 138(5): 961-75, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19737522

ABSTRACT

Obesity is associated with chronic low-grade inflammation that negatively impacts insulin sensitivity. Here, we show that high-fat diet can increase NF-kappaB activation in mice, which leads to a sustained elevation in level of IkappaB kinase epsilon (IKKepsilon) in liver, adipocytes, and adipose tissue macrophages. IKKepsilon knockout mice are protected from high-fat diet-induced obesity, chronic inflammation in liver and fat, hepatic steatosis, and whole-body insulin resistance. These mice show increased energy expenditure and thermogenesis via enhanced expression of the uncoupling protein UCP1. They maintain insulin sensitivity in liver and fat, without activation of the proinflammatory JNK pathway. Gene expression analyses indicate that IKKepsilon knockout reduces expression of inflammatory cytokines, and changes expression of certain regulatory proteins and enzymes involved in glucose and lipid metabolism. Thus, IKKepsilon may represent an attractive therapeutic target for obesity, insulin resistance, diabetes, and other complications associated with these disorders.


Subject(s)
Energy Metabolism , I-kappa B Kinase/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Fatty Liver , I-kappa B Kinase/genetics , Insulin/metabolism , Insulin Resistance , Lipid Metabolism , Liver/metabolism , Mice , Mice, Knockout , Mice, Transgenic , NF-kappa B/metabolism , Obesity/immunology
5.
EMBO J ; 37(1): 19-38, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29150432

ABSTRACT

The innate immune kinase TBK1 initiates inflammatory responses to combat infectious pathogens by driving production of type I interferons. TBK1 also controls metabolic processes and promotes oncogene-induced cell proliferation and survival. Here, we demonstrate that TBK1 activates mTOR complex 1 (mTORC1) directly. In cultured cells, TBK1 associates with and activates mTORC1 through site-specific mTOR phosphorylation (on S2159) in response to certain growth factor receptors (i.e., EGF-receptor but not insulin receptor) and pathogen recognition receptors (PRRs) (i.e., TLR3; TLR4), revealing a stimulus-selective role for TBK1 in mTORC1 regulation. By studying cultured macrophages and those isolated from genome edited mTOR S2159A knock-in mice, we show that mTOR S2159 phosphorylation promotes mTORC1 signaling, IRF3 nuclear translocation, and IFN-ß production. These data demonstrate a direct mechanistic link between TBK1 and mTORC1 function as well as physiologic significance of the TBK1-mTORC1 axis in control of innate immune function. These data unveil TBK1 as a direct mTORC1 activator and suggest unanticipated roles for mTORC1 downstream of TBK1 in control of innate immunity, tumorigenesis, and disorders linked to chronic inflammation.


Subject(s)
Immunity, Innate/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cytosol/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Macrophages/drug effects , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Transport , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
6.
Am J Physiol Heart Circ Physiol ; 320(1): H323-H337, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33164548

ABSTRACT

Interleukin-4 receptor α (IL4Rα) signaling plays an important role in cardiac remodeling during myocardial infarction (MI). However, the target cell type(s) of IL4Rα signaling during this remodeling remains unclear. Here, we investigated the contribution of endogenous myeloid-specific IL4Rα signaling in cardiac remodeling post-MI. We established a murine myeloid-specific IL4Rα knockout (MyIL4RαKO) model with LysM promoter-driven Cre recombination. Macrophages from MyIL4RαKO mice showed significant downregulation of alternatively activated macrophage markers but an upregulation of classical activated macrophage markers both in vitro and in vivo, indicating the successful inactivation of IL4Rα signaling in macrophages. To examine the role of myeloid IL4Rα during MI, we subjected MyIL4RαKO and littermate floxed control (FC) mice to MI. We found that cardiac function was significantly impaired as a result of myeloid-specific IL4Rα deficiency. This deficiency resulted in a dysregulated inflammatory response consisting of decreased production of anti-inflammatory cytokines. Myeloid IL4Rα deficiency also led to reduced collagen 1 deposition and an imbalance of matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs), with upregulated MMPs and downregulated TIMPs, which resulted in insufficient fibrotic remodeling. In conclusion, this study identifies that myeloid-specific IL4Rα signaling regulates inflammation and fibrotic remodeling during MI. Therefore, myeloid-specific activation of IL4Rα signaling could offer protective benefits after MI.NEW & NOTEWORTHY This study showed, for the first time, the role of endogenous IL4Rα signaling in myeloid cells during cardiac remodeling and the underlying mechanisms. We identified myeloid cells are the critical target cell types of IL4Rα signaling during cardiac remodeling post-MI. Deficiency of myeloid IL4Rα signaling causes deteriorated cardiac function post-MI, due to dysregulated inflammation and insufficient fibrotic remodeling. This study sheds light on the potential of activating myeloid-specific IL4Rα signaling to modify remodeling post-MI. This brings hope to patients with MI and diminishes side effects by cell type-specific instead of whole body treatment.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Receptors, Cell Surface/metabolism , Ventricular Function, Left , Ventricular Remodeling , Animals , Cells, Cultured , Disease Models, Animal , Fibrosis , Macrophage Activation , Macrophages/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction
7.
J Immunol ; 202(3): 931-942, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30578307

ABSTRACT

Sepsis is the leading cause of death in the intensive care unit with an overall mortality rate of 20%. Individuals who are obese and have type 2 diabetes have increased recurrent, chronic, nosocomial infections that worsen the long-term morbidity and mortality from sepsis. Additionally, animal models of sepsis have shown that obese, diabetic mice have lower survival rates compared with nondiabetic mice. Neutrophils are essential for eradication of bacteria, prevention of infectious complications, and sepsis survival. In diabetic states, there is a reduction in neutrophil chemotaxis, phagocytosis, and reactive oxygen species (ROS) generation; however, few studies have investigated the extent to which these deficits compromise infection eradication and mortality. Using a cecal ligation and puncture model of sepsis in lean and in diet-induced obese mice, we demonstrate that obese diabetic mice have decreased "emergency hematopoiesis" after an acute infection. Additionally, both neutrophils and monocytes in obese, diabetic mice have functional defects, with decreased phagocytic ability and a decreased capacity to generate ROS. Neutrophils isolated from obese diabetic mice have decreased transcripts of Axl and Mertk, which partially explains the phagocytic dysfunction. Furthermore, we found that exogenous GM-CSF administration improves sepsis survival through enhanced neutrophil and monocytes phagocytosis and ROS generation abilities in obese, diabetic mice with sepsis.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Immunity, Innate/drug effects , Obesity/immunology , Sepsis/immunology , Animals , Bacteria , Cytokines/genetics , Cytokines/immunology , Diabetes Mellitus, Experimental/microbiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/pathology , Neutrophils/immunology , Neutrophils/pathology , Obesity/microbiology , Phagocytosis , Sepsis/drug therapy , Sepsis/microbiology
8.
Biotechnol Bioeng ; 117(12): 3891-3901, 2020 12.
Article in English | MEDLINE | ID: mdl-32729936

ABSTRACT

Dysfunctional adipose tissue plays a central role in the pathogenesis of the obesity-related metabolic disease, including type 2 diabetes. Targeting adipose tissue using biopolymer implants is a novel therapeutic approach for metabolic disease. We transplanted porous poly(lactide-co-glycolide) (PLG) implants coated with human interleukin-4 (hIL-4)-expressing lentivirus into epididymal white adipose tissue (eWAT) of mice fed a high-fat diet. Tissue and systemic inflammation and metabolism were studied with flow cytometry, immunohistochemistry, quantitative real-time polymerase chain reaction, adipose tissue histology, and in vivo glucose tolerance testing at 2 and 10 weeks of a high-fat diet. PLG implants carrying hIL-4-expressing lentivirus implanted into epididymal white adipose tissue of mice-regulated adipose tissue inflammation, including increased CD3+ CD4+ T-cell frequency, increased eWAT adipocyte hypertrophy, and decreased FASN and ATGL expression, along with reduced fasting blood glucose levels. These effects were observed in early obesity but were not maintained in established obesity. Local delivery of bioimplants loaded with cytokine-expressing lentivirus vectors to adipose tissue influences tissue inflammation and systemic metabolism in early obesity. Further study will be required to show more durable metabolic effects. These data demonstrate that polymer biomaterials implanted into adipose tissue have the potential to modulate local tissue and systemic inflammation and metabolism.


Subject(s)
Adipose Tissue/metabolism , Implants, Experimental , Interleukin-4 , Lentivirus , Obesity/metabolism , Transduction, Genetic , Animals , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/biosynthesis , Interleukin-4/genetics , Male , Mice , Obesity/genetics
9.
J Biol Chem ; 293(23): 8775-8786, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29636416

ABSTRACT

Obesity-induced chronic inflammation is associated with metabolic disease. Results from mouse models utilizing a high-fat diet (HFD) have indicated that an increase in activated macrophages, including CD11c+ adipose tissue macrophages (ATMs), contributes to insulin resistance. Obesity primes myeloid cell production from hematopoietic stem cells (HSCs) and Toll-like receptor 4 (TLR4), and the downstream TIR domain-containing adapter protein-inducing interferon-ß (TRIF)- and MyD88-mediated pathways regulate production of similar myeloid cells after lipopolysaccharide stimulation. However, the role of these pathways in HFD-induced myelopoiesis is unknown. We hypothesized that saturated fatty acids and HFD alter myelopoiesis by activating TLR4 pathways in HSCs, differentially producing pro-inflammatory CD11c+ myeloid cells that contribute to obesity-induced metabolic disease. Results from reciprocal bone marrow transplants (BMTs) with Tlr4-/- and WT mice indicated that TLR4 is required for HFD-induced myelopoiesis and production of CD11c+ ATMs. Experiments with homozygous knockouts of Irakm (encoding a suppressor of MyD88 inactivation) and Trif in competitive BMTs revealed that MyD88 is required for HFD expansion of granulocyte macrophage progenitors and that Trif is required for pregranulocyte macrophage progenitor expansion. A comparison of WT, Tlr4-/-, Myd88-/-, and Trif-/- mice on HFD demonstrated that TLR4 plays a role in the production of CD11c+ ATMs, and both Myd88-/- and Trif-/- mice produced fewer ATMs than WT mice. Moreover, HFD-induced TLR4 activation inhibited macrophage proliferation, leading to greater accumulation of recruited CD11c+ ATMs. Our results indicate that HFD potentiates TLR4 and both its MyD88- and TRIF-mediated downstream pathways within progenitors and adipose tissue and leads to macrophage polarization.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , CD11c Antigen/immunology , Macrophages/pathology , Myeloid Differentiation Factor 88/immunology , Myelopoiesis , Obesity/pathology , Toll-Like Receptor 4/immunology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/etiology , Obesity/immunology
10.
Immunology ; 155(4): 407-417, 2018 12.
Article in English | MEDLINE | ID: mdl-30229891

ABSTRACT

The expansion of adipose tissue (AT) in obesity is accompanied by the accumulation of immune cells that contribute to a state of low-grade, chronic inflammation and dysregulated metabolism. Adipose tissue macrophages (ATMs) represent the most abundant class of leukocytes in AT and are involved in the regulation of several regulatory physiological processes, such as tissue remodeling and insulin sensitivity. With progressive obesity, ATMs are key mediators of meta-inflammation, insulin resistance and impairment of adipocyte function. While macrophage recruitment from blood monocytes is a critical component of the generation of AT inflammation, new studies have revealed a role for ATM proliferation in the early stages of obesity and in sustaining AT inflammation. In addition, studies have revealed a more complex range of macrophage activation states than the previous M1/M2 model, and the existence of different macrophage profiles between human and animal models. This review will summarize the current understanding of the regulatory mechanisms of ATM function in relation to obesity, type 2 diabetes, depot of origin, and to other leukocytes such as AT dendritic cells, with hopes of emphasizing the regulatory nodes that can potentially be targeted to prevent and treat obesity-related metabolic disorders.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/immunology , Macrophages/immunology , Obesity/immunology , Obesity/pathology , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Diabetes Mellitus, Type 2/immunology , Humans , Inflammation/immunology , Insulin Resistance/immunology , Macrophage Activation/immunology , Mice
11.
J Immunol ; 197(9): 3650-3661, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27683748

ABSTRACT

Dynamic changes of adipose tissue leukocytes, including adipose tissue macrophage (ATM) and adipose tissue dendritic cells (ATDCs), contribute to obesity-induced inflammation and metabolic disease. However, clear discrimination between ATDC and ATM in adipose tissue has limited progress in the field of immunometabolism. In this study, we use CD64 to distinguish ATM and ATDC, and investigated the temporal and functional changes in these myeloid populations during obesity. Flow cytometry and immunostaining demonstrated that the definition of ATM as F4/80+CD11b+ cells overlaps with other leukocytes and that CD45+CD64+ is specific for ATM. The expression of core dendritic cell genes was enriched in CD11c+CD64- cells (ATDC), whereas core macrophage genes were enriched in CD45+CD64+ cells (ATM). CD11c+CD64- ATDCs expressed MHC class II and costimulatory receptors, and had similar capacity to stimulate CD4+ T cell proliferation as ATMs. ATDCs were predominantly CD11b+ conventional dendritic cells and made up the bulk of CD11c+ cells in adipose tissue with moderate high-fat diet exposure. Mixed chimeric experiments with Ccr2-/- mice demonstrated that high-fat diet-induced ATM accumulation from monocytes was dependent on CCR2, whereas ATDC accumulation was less CCR2 dependent. ATDC accumulation during obesity was attenuated in Ccr7-/- mice and was associated with decreased adipose tissue inflammation and insulin resistance. CD45+CD64+ ATM and CD45+CD64-CD11c+ ATDCs were identified in human obese adipose tissue and ATDCs were increased in s.c. adipose tissue compared with omental adipose tissue. These results support a revised strategy for unambiguous delineation of ATM and ATDC, and suggest that ATDCs are independent contributors to adipose tissue inflammation during obesity.


Subject(s)
Adipose Tissue/immunology , Dendritic Cells/immunology , Inflammation/immunology , Macrophages/immunology , Obesity/immunology , Animals , Cells, Cultured , Diet, High-Fat , Gene Expression Profiling , Humans , Immunophenotyping , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR7/genetics , Receptors, IgG/metabolism
12.
J Biol Chem ; 290(21): 13250-62, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25869128

ABSTRACT

Women of reproductive age are protected from metabolic disease relative to postmenopausal women and men. Most preclinical rodent studies are skewed toward the use of male mice to study obesity-induced metabolic dysfunction because of a similar protection observed in female mice. How sex differences in obesity-induced inflammatory responses contribute to these observations is unknown. We have compared and contrasted the effects of high fat diet-induced obesity on glucose metabolism and leukocyte activation in multiple depots in male and female C57Bl/6 mice. With both short term and long term high fat diet, male mice demonstrated increased weight gain and CD11c(+) adipose tissue macrophage content compared with female mice despite similar degrees of adipocyte hypertrophy. Competitive bone marrow transplant studies demonstrated that obesity induced a preferential contribution of male hematopoietic cells to circulating leukocytes and adipose tissue macrophages compared with female cells independent of the sex of the recipient. Sex differences in macrophage and hematopoietic cell in vitro activation in response to obesogenic cues were observed to explain these results. In summary, this report demonstrates that male and female leukocytes and hematopoietic stem cells have cell-autonomous differences in their response to obesity that contribute to an amplified response in males compared with females.


Subject(s)
Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Hematopoietic Stem Cells/cytology , Inflammation/immunology , Obesity/etiology , Adipose Tissue/cytology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Biomarkers/analysis , Cells, Cultured , Colony-Forming Units Assay , Female , Flow Cytometry , Glucose Tolerance Test , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Inflammation/complications , Inflammation/pathology , Lipids/analysis , Male , Mice , Mice, Inbred C57BL , Myelopoiesis/physiology , Obesity/metabolism , Obesity/pathology , Sex Factors , Weight Gain
13.
J Cell Sci ; 126(Pt 8): 1733-43, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444381

ABSTRACT

Previous studies have shown that growth hormone (GH) recruits the adapter protein SH2B1ß to the GH-activated, GH receptor-associated tyrosine kinase JAK2, implicating SH2B1ß in GH-dependent actin cytoskeleton remodeling, and suggesting that phosphorylation at serines 161 and 165 in SH2B1ß releases SH2B1ß from the plasma membrane. Here, we examined the role of SH2B1ß in GH regulation of macrophage migration. We show that GH stimulates migration of cultured RAW264.7 macrophages, and primary cultures of peritoneal and bone marrow-derived macrophages. SH2B1ß overexpression enhances, whereas SH2B1 knockdown inhibits, GH-dependent motility of RAW macrophages. At least two independent mechanisms regulate the SH2B1ß-mediated changes in motility. In response to GH, tyrosines 439 and 494 in SH2B1ß are phosphorylated. Mutating these tyrosines in SH2B1ß decreases both basal and GH-stimulated macrophage migration. In addition, mutating the polybasic nuclear localization sequence (NLS) in SH2B1ß or creating the phosphomimetics SH2B1ß(S161E) or SH2B1ß(S165E), all of which release SH2B1ß from the plasma membrane, enhances macrophage motility. Conversely, SH2B1ß(S161/165A) exhibits increased localization at the plasma membrane and decreased macrophage migration. Mutating the NLS or the nearby serine residues does not alter GH-dependent phosphorylation on tyrosines 439 and 494 in SH2B1ß. Mutating tyrosines 439 and 494 does not affect localization of SH2B1ß at the plasma membrane or movement of SH2B1ß into focal adhesions. Taken together, these results suggest that SH2B1ß enhances GH-stimulated macrophage motility via mechanisms involving phosphorylation of SH2B1ß on tyrosines 439 and 494 and movement of SH2B1ß out of the plasma membrane (e.g. as a result of phosphorylation of serines 161 and 165).


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Growth Hormone/metabolism , Macrophages/cytology , Macrophages/metabolism , Animals , Cell Line , Cell Movement/physiology , Focal Adhesions/metabolism , Mice , Phosphorylation
14.
Ergonomics ; 58(2): 301-9, 2015.
Article in English | MEDLINE | ID: mdl-25323820

ABSTRACT

We present a new method for rapidly measuring child body shapes from noisy, incomplete data captured from low-cost depth cameras. This method fits the data using a statistical body shape model (SBSM) to find a complete avatar in the realistic body shape space. The method also predicts a set of standard anthropometric data for a specific subject without measuring dimensions directly from the fitted model. Since the SBSM was developed using principal component (PC) analysis, we formulate an optimisation problem to fit the model in which the degrees of freedom are defined in PC-score space. The mean unsigned distance between the fitted-model based on depth-camera data and the high-resolution laser scan data was 9.4 mm with a standard deviation (SD) of 5.1 mm. For the torso, the mean distance was 2.9 mm (SD 1.4 mm). The correlations between standard anthropometric dimensions predicted by the SBSM and manually measured dimensions exceeded 0.9.


Subject(s)
Anthropometry/instrumentation , Body Size , Imaging, Three-Dimensional/instrumentation , Models, Statistical , Photography/instrumentation , Anthropometry/methods , Child , Child, Preschool , Female , Humans , Imaging, Three-Dimensional/methods , Lasers , Male , Photography/methods , Principal Component Analysis
15.
J Biol Chem ; 288(22): 15725-35, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23595986

ABSTRACT

We investigated GH action on macrophage (MΦ) by creating a MΦ-specific GH receptor-null mouse model (MacGHR KO). On a normal diet (10% fat), MacGHR KO and littermate controls exhibited similar growth profiles and glucose excursions on intraperitoneal glucose (ipGTT) and insulin tolerance (ITT) tests. However, when challenged with high fat diet (HFD, 45% fat) for 18 weeks, MacGHR KO mice exhibited impaired ipGTT and ITT compared with controls. In MacGHR KO, adipose-tissue (AT) MΦ abundance was increased with skewing toward M1 polarization. Expression of pro-inflammatory cytokines (IL1ß, TNF-α, IL6, and osteopontin (OPN)) were increased in MacGHR KO AT stromal vascular fraction (SVF). In MacGHR KO AT, crown-like-structures were increased with decreased insulin-dependent Akt phosphorylation. The abundance of phosphorylated NF-κB and of OPN was increased in SVF and bone-marrow-derived MΦ in MacGHR KO. GH, acting via an NF-κB site in the distal OPN promoter, inhibited the OPN promoter. Thus in diet-induced obesity (DIO), lack of GH action on the MΦ exerts an unexpected deleterious effect on glucose homeostasis by accentuating AT inflammation and NF-κB-dependent activation of OPN expression. These novel results in mice support the possibility that administration of GH could have salutary effects on DIO-associated chronic inflammation and insulin resistance in humans.


Subject(s)
Carrier Proteins/metabolism , Diet/adverse effects , Glucose/metabolism , Growth Hormone/metabolism , Homeostasis , Insulin Resistance , Macrophages, Peritoneal/metabolism , Obesity/metabolism , Osteopontin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Carrier Proteins/genetics , Glucose/genetics , Growth Hormone/genetics , Humans , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Osteopontin/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Response Elements/genetics
16.
Nat Med ; 13(4): 455-62, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17351624

ABSTRACT

Cbl-associated protein (Cap) is a member of a phosphatidylinositol 3-kinase-independent pathway for insulin-stimulated translocation of the glucose transporter GLUT4. Despite this positive role of Cap in glucose uptake, here we show that deletion of the gene encoding Cap (official gene name: Sorbs1) protects against high-fat diet (HFD)-induced insulin resistance in mice while also having an opposite, insulin-sensitizing effect, accompanied by reduced tissue markers of inflammation. Given the emerging role of chronic inflammation in insulin resistance and the macrophage in initiating this inflammatory process, we considered that Sorbs1 deletion from macrophages may have resulted in the observed protection from HFD-induced insulin resistance. Using bone marrow transplantation to generate functional Sorbs1-null macrophages, we show that the insulin-sensitive phenotype can be transferred to wild-type mice by transplantation of Sorbs1-null bone marrow. These studies show that macrophages are an important cell type in the induction of insulin resistance and that Cap has a modulatory role in this function.


Subject(s)
Gene Deletion , Insulin Resistance/genetics , Microfilament Proteins/genetics , Adipocytes/metabolism , Animals , Bone Marrow Transplantation , Dietary Fats , Histocytochemistry , Immunoblotting , Insulin Resistance/physiology , Macrophages/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Microfilament Proteins/metabolism
17.
Nat Rev Endocrinol ; 20(1): 50-61, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37872302

ABSTRACT

The signals and structure of the tissues in which leukocytes reside critically mould leukocyte function and development and have challenged our fundamental understanding of how to define and categorize tissue-resident immune cells. One specialized tissue niche that has a powerful effect on immune cell function is adipose tissue. The field of adipose tissue leukocyte biology has expanded dramatically and has revealed how tissue niches can shape immune cell function and reshape them in a setting of metabolic stress, such as obesity. Most notably, adipose tissue macrophages and T cells are under intense investigation due to their contributions to adipose tissue in the lean and obese states. Both adipose tissue macrophages and T cells have features associated with the metabolic function of adipose tissue that are distinct from features of macrophages and T cells that are classically characterized in other tissues. This Review provides state-of-the-art understanding of adipose tissue macrophages and T cells and discusses how their unique niche can help us to better understand diversity in leukocyte responses.


Subject(s)
Adipose Tissue , T-Lymphocytes , Humans , Adipose Tissue/metabolism , Macrophages , Obesity/metabolism , Inflammation/metabolism
18.
J Immunol ; 187(12): 6208-16, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22075699

ABSTRACT

Age-related adiposity has been linked to chronic inflammatory diseases in late life. To date, the studies on adipose tissue leukocytes and aging have not taken into account the heterogeneity of adipose tissue macrophages (ATMs), nor have they examined how age impacts other leukocytes such as T cells in fat. Therefore, we have performed a detailed examination of ATM subtypes in young and old mice using state of the art techniques. Our results demonstrate qualitative changes in ATMs with aging that generate a decrease in resident type 2 (M2) ATMs. The profile of ATMs in old fat shifts toward a proinflammatory environment with increased numbers of CD206(-)CD11c(-) (double-negative) ATMs. The mechanism of this aging-induced shift in the phenotypic profile of ATMs was found to be related to a decrease in peroxisome proliferator-activated receptor-γ expression in ATMs and alterations in chemokine/chemokine receptor expression profiles. Furthermore, we have revealed a profound and unexpected expansion of adipose tissue T cells in visceral fat with aging that includes a significant induction of regulatory T cells in fat. Our findings demonstrate a unique inflammatory cell signature in the physiologic context of aging adipose tissue that differs from those induced in setting of diet-induced obesity.


Subject(s)
Aging/immunology , Aging/pathology , Cellular Senescence/immunology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Leukocytes, Mononuclear/immunology , Animals , Cell Separation/methods , Immunophenotyping , Inflammation/immunology , Inflammation/pathology , Intra-Abdominal Fat/cytology , Leukocyte Count , Leukocytes, Mononuclear/classification , Leukocytes, Mononuclear/pathology , Lymphocyte Count , Macrophages, Peritoneal/classification , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology , Spleen/cytology , Spleen/immunology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
19.
Sci Rep ; 13(1): 2651, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36788340

ABSTRACT

Obesity induces a chronic inflammatory state associated with changes in adipose tissue macrophages (ATMs). Macrophage scavenger receptor 1 (MSR1) has been implicated in the regulation of adipose tissue inflammation and diabetes pathogenesis; however, reports have been mixed on the contribution of MSR1 in obesity and glucose intolerance. We observed increased MSR1 expression in VAT of obese diabetic individuals compared to non-diabetic and single nuclear RNA sequencing identified macrophage-specific expression of MSR1 in human adipose tissue. We examined male Msr1-/- (Msr1KO) and WT controls and observed protection from obesity and AT inflammation in non-littermate Msr1KO mice. We then evaluated obese littermate Msr1+/- (Msr1HET) and Msr1KO mice. Both Msr1KO mice and Msr1HET mice became obese and insulin resistant when compared to their normal chow diet counterparts, but there was no Msr1-dependent difference in body weight, glucose metabolism, or insulin resistance. Flow cytometry revealed no significant differences between genotypes in ATM subtypes or proliferation in male and female mice. We observed increased frequency of proliferating ATMs in obese female compared to male mice. Overall, we conclude that while MSR1 is a biomarker of diabetes status in human adipose tissue, in mice Msr1 is not required for obesity-associated insulin resistance or ATM accumulation.


Subject(s)
Insulin Resistance , Obesity , Scavenger Receptors, Class A , Animals , Female , Male , Mice , Adipose Tissue/metabolism , Inflammation/metabolism , Insulin/metabolism , Insulin Resistance/genetics , Mice, Inbred C57BL , Obesity/complications , Obesity/genetics , Obesity/metabolism , Scavenger Receptors, Class A/metabolism
20.
Adipocyte ; 12(1): 2268261, 2023 12.
Article in English | MEDLINE | ID: mdl-37815174

ABSTRACT

Alterations of the extracellular matrix contribute to adipose tissue dysfunction in metabolic disease. We studied the role of matrix density in regulating human adipocyte phenotype in a tunable hydrogel culture system. Lipid accumulation was maximal in intermediate hydrogel density of 5 weight %, relative to 3% and 10%. Adipogenesis and lipid and oxidative metabolic gene pathways were enriched in adipocytes in 5% relative to 3% hydrogels, while fibrotic gene pathways were enriched in 3% hydrogels. These data demonstrate that the intermediate density matrix promotes a more adipogenic, less fibrotic adipocyte phenotype geared towards increased lipid and aerobic metabolism. These observations contribute to a growing literature describing the role of matrix density in regulating adipose tissue function.


Subject(s)
Adipocytes , Adipose Tissue , Humans , Adipocytes/metabolism , Adipogenesis/genetics , Hydrogels/metabolism , Phenotype , Lipids
SELECTION OF CITATIONS
SEARCH DETAIL