Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Rev Endocrinol ; 19(12): 691-707, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37749386

ABSTRACT

Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.


Subject(s)
Adipose Tissue , Obesity , Humans , Adipose Tissue/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adipokines/metabolism , Biology
2.
Biochem Mol Biol Educ ; 51(5): 508-519, 2023.
Article in English | MEDLINE | ID: mdl-37354049

ABSTRACT

Graduate programs in medicine and biomedical sciences have been severely impacted by the SARS-CoV-2/COVID-19 pandemic over the last 2 years. Following 2 years since beginning of the pandemic, data on student support, educational and academic performance as well as sentiment on changes to educational programs are starting to emerge. We performed and compared results of two cross-sectional surveys of Swedish and U.S.-based medical and biomedical graduate students on how the pandemic has affected their studies, research productivity and career trajectory. Students were also asked to assess support provided by the university and supervisors. The surveys also captured student demographics and a range of other factors, such as pressures brought on by caretaking and financial responsibilities. We analyzed answers from 264 and 106 students attending graduate programs in universities in Sweden and the United States, respectively. U.S.-based students faced more severe restrictions on their research program compared to students in Sweden, reporting more delays in productivity, scientific output and graduation, and increased worries about their career trajectory. Swedish students had more caretaking responsibilities, although these did not cause any delays in graduation. While support by universities and supervisors was comparable between the countries, financial worries and mental health concerns were particularly prominent in the U.S. cohort. Student performance and outlook was hugely dependent on the breadth of the restrictions and the available support. Besides the governmental and university-led approach to counter the pandemic, societal differences also played a role in how well students were handling effects of the pandemic.


Subject(s)
COVID-19 , Humans , United States/epidemiology , Cross-Sectional Studies , Sweden/epidemiology , COVID-19/epidemiology , Pandemics , SARS-CoV-2 , Students
3.
Front Cell Dev Biol ; 10: 1003118, 2022.
Article in English | MEDLINE | ID: mdl-36187476

ABSTRACT

The prevalence of obesity and metabolic diseases continues to rise, which has led to an increased interest in studying adipose tissue to elucidate underlying disease mechanisms. The use of genetic mouse models has been critical for understanding the role of specific genes for adipose tissue function and the tissue's impact on other organs. However, mouse adipose tissue displays key differences to human fat, which has led, in some cases, to the emergence of some confounding concepts in the adipose field. Such differences include the depot-specific characteristics of visceral and subcutaneous fat, and divergences in thermogenic fat phenotype between the species. Adipose tissue characteristics may therefore not always be directly compared between species, which is important to consider when setting up new studies or interpreting results. This mini review outlines our current knowledge about the cell biological differences between human and mouse adipocytes and fat depots, highlighting some examples where inadequate knowledge of species-specific differences can lead to confounding results, and presenting plausible anatomic explanations that may underlie the differences. The article thus provides critical insights and guidance for researchers working primarily with only human or mouse fat tissue, and may contribute to new ideas or concepts in the important and evolving field of adipose biology.

4.
iScience ; 25(7): 104602, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35789845

ABSTRACT

Adipose tissue inflammation drives obesity-related cardiometabolic diseases. Enhancing endogenous resolution mechanisms through administration of lipoxin A4, a specialized pro-resolving lipid mediator, was shown to reduce adipose inflammation and subsequently protects against obesity-induced systemic disease in mice. Here, we demonstrate that lipoxins reduce inflammation in 3D-cultured human adipocytes and adipose tissue explants from obese patients. Approximately 50% of patients responded particularly well to lipoxins by reducing inflammatory cytokines and promoting an anti-inflammatory M2 macrophage phenotype. Responding patients were characterized by elevated systemic levels of C-reactive protein, which causes inflammation in cultured human adipocytes. Responders appeared more prone to producing anti-inflammatory oxylipins and displayed elevated prostaglandin D2 levels, which has been interlinked with transcription of lipoxin-generating enzymes. Using explant cultures, this study provides the first proof-of-concept evidence supporting the therapeutic potential of lipoxins in reducing human adipose tissue inflammation. Our data further indicate that lipoxin treatment may require a tailored personalized-medicine approach.

5.
Sci Adv ; 8(17): eabm7012, 2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35476450

ABSTRACT

The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.

6.
FASEB J ; 36(3): e22173, 2022 03.
Article in English | MEDLINE | ID: mdl-35104001

ABSTRACT

Dysregulated chronic inflammation plays a crucial role in the pathophysiology of atherosclerosis and may be a result of impaired resolution. Thus, restoring levels of specialized pro-resolving mediators (SPMs) to promote the resolution of inflammation has been proposed as a therapeutic strategy for patients with atherosclerosis, in addition to standard clinical care. Herein, we evaluated the effects of the SPM lipids, lipoxin A4 (LXA4 ) and lipoxin B4 (LXB4 ), on neutrophils isolated from patients with atherosclerosis compared with healthy controls. Patients displayed altered endogenous SPM production, and we demonstrated that lipoxin treatment in whole blood from atherosclerosis patients attenuates neutrophil oxidative burst, a key contributor to atherosclerotic development. We found the opposite effect in neutrophils from healthy controls, indicating a potential mechanism whereby lipoxins aid the endogenous neutrophil function in health but reduce its excessive activation in disease. We also demonstrated that lipoxins attenuated upregulation of the high-affinity conformation of the CD11b/CD18 integrin, which plays a central role in clot activation and atherosclerosis. Finally, LXB4 enhanced lymphatic transmigration of human neutrophils isolated from patients with atherosclerosis. This finding is noteworthy, as impaired lymphatic function is now recognized as an important contributor to atherosclerosis. Although both lipoxins modulated neutrophil function, LXB4 displayed more potent effects than LXA4 in humans. This study highlights the therapeutic potential of lipoxins in atherosclerotic disease and demonstrates that the effect of these SPMs may be specifically tailored to the need of the individual.


Subject(s)
Atherosclerosis/metabolism , Integrins/metabolism , Lipoxins/metabolism , Neutrophils/metabolism , Respiratory Burst/physiology , Aged , Female , Humans , Inflammation/metabolism , Male , Middle Aged
7.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008946

ABSTRACT

Obesity is associated with extensive expansion and remodeling of the adipose tissue architecture, including its microenvironment and extracellular matrix (ECM). Although obesity has been reported to induce adipose tissue fibrosis, the composition of the ECM under healthy physiological conditions has remained underexplored and debated. Here, we used a combination of three established techniques (picrosirius red staining, a colorimetric hydroxyproline assay, and sensitive gene expression measurements) to evaluate the status of the ECM in metabolically healthy lean (MHL) and metabolically unhealthy obese (MUO) subjects. We investigated ECM deposition in the two major human adipose tissues, namely the omental and subcutaneous depots. Biopsies were obtained from the same anatomic region of respective individuals. We found robust ECM deposition in MHL subjects, which correlated with high expression of collagens and enzymes involved in ECM remodeling. In contrast, MUO individuals showed lower expression of ECM components but elevated levels of ECM cross-linking and adhesion proteins, e.g., lysyl oxidase and thrombospondin. Our data suggests that subcutaneous fat is more prone to express proteins involved in ECM remodeling than omental adipose tissues. We conclude that a more dynamic ability to deposit and remodel ECM may be a key signature of healthy adipose tissue, and that subcutaneous fat may adapt more readily to changing metabolic conditions than omental fat.


Subject(s)
Adipose Tissue/metabolism , Extracellular Matrix/metabolism , Gene Expression , Omentum/metabolism , Subcutaneous Fat/metabolism , Adult , Biomarkers , Collagen/metabolism , Female , Humans , Male , Middle Aged , Organ Specificity/genetics , RNA, Messenger/genetics , Sensitivity and Specificity
8.
BMC Med Educ ; 21(1): 294, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022871

ABSTRACT

BACKGROUND: It remains unclear to what extent the SARS-CoV-2/COVID-19 pandemic disrupted the normal progression of biomedical and medical science graduate programs and if there was a lasting impact on the quality and quantity of supervision of PhD-students. To date, multiple editorials and commentaries indicate the severity of the disruption without providing sufficient evidence with quantifiable data. METHODS: An online survey was submitted to the administrative offices of biomedical and medical PhD-programs at eight major universities in Sweden to gauge the impact of the pandemic on the students. It consisted of multiple-choice and open-ended questions where students could provide examples of positive and/or negative supervision strategies. Open answered questions were coded as either examples of positive or negative support. RESULTS: PhD students were divided into two groups: those with improved or unchanged supervision during the pandemic (group 1, n = 185), versus those whose supervision worsened (group 2, n = 69). Group 1 received more help from supervisors and more frequent supervision via both online and alternative platforms (email/messages and telephone). There was no significant difference in educational-stage, gender or caretaking responsibilities between the groups. CONCLUSIONS: It is important for the scientific community to learn how to provide the best possible supervision for PhD students during the pandemic. Our data suggests that more frequent supervision, and using a diverse array of meeting platforms is helpful. In addition, it is important for the students to feel that they have their supervisor's emotional support. Several students also expressed that they would benefit from an extension of their PhD programs due to delays caused by the pandemic.


Subject(s)
COVID-19 , Pandemics , Cross-Sectional Studies , Education, Graduate , Humans , SARS-CoV-2 , Students , Sweden/epidemiology
9.
Int J Mol Sci ; 22(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803130

ABSTRACT

Diminished lymphatic function and abnormal morphology are common in chronic inflammatory diseases. Recent studies are investigating whether it is possible to target chronic inflammation by promoting resolution of inflammation, in order to enhance lymphatic function and attenuate disease. Resolution of inflammation is an active process regulated by bioactive lipids known as specialized pro-resolving mediators (SPMs). SPMs can modulate leukocyte migration and function, alter cytokine/chemokine release, modify autophagy, among other immune-related activities. Here, we summarize the role of the lymphatics in resolution of inflammation and lymphatic impairment in chronic inflammatory diseases. Furthermore, we discuss the current literature describing the connection between SPMs and the lymphatics, and the possibility of targeting the lymphatics with innovative SPM therapy to promote resolution of inflammation and mitigate disease.


Subject(s)
Cell Movement , Chemokines/metabolism , Inflammation Mediators/metabolism , Leukocytes/metabolism , Lymphatic System/metabolism , Animals , Chronic Disease , Humans , Inflammation/metabolism , Inflammation/pathology , Leukocytes/pathology , Lymphatic System/pathology
10.
J Clin Med ; 10(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801198

ABSTRACT

Three members of the obscurin protein family that contain tandem kinase domains with important signaling functions for cardiac and striated muscles are the giant protein obscurin, its obscurin-associated kinase splice isoform, and the striated muscle enriched protein kinase (SPEG). While there is increasing evidence for the specific roles that each individual kinase domain plays in cross-striated muscles, their biology and regulation remains enigmatic. Our present study focuses on kinase domain 1 and the adjacent low sequence complexity inter-kinase domain linker in obscurin and SPEG. Using Phos-tag gels, we show that the linker in obscurin contains several phosphorylation sites, while the same region in SPEG remained unphosphorylated. Our homology modeling, mutational analysis and molecular docking demonstrate that kinase 1 in obscurin harbors all key amino acids important for its catalytic function and that actions of this domain result in autophosphorylation of the protein. Our bioinformatics analyses also assign a list of putative substrates for kinase domain 1 in obscurin and SPEG, based on the known and our newly proposed phosphorylation sites in muscle proteins, including obscurin itself.

11.
J Gen Physiol ; 153(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-33836065

ABSTRACT

The N2A segment of titin is a main signaling hub in the sarcomeric I-band that recruits various signaling factors and processing enzymes. It has also been proposed to play a role in force production through its Ca2+-regulated association with actin. However, the molecular basis by which N2A performs these functions selectively within the repetitive and extensive titin chain remains poorly understood. Here, we analyze the structure of N2A components and their association with F-actin. Specifically, we characterized the structure of its Ig domains by elucidating the atomic structure of the I81-I83 tandem using x-ray crystallography and computing a homology model for I80. Structural data revealed these domains to present heterogeneous and divergent Ig folds, where I81 and I83 have unique loop structures. Notably, the I81-I83 tandem has a distinct rotational chain arrangement that confers it a unique multi-domain topography. However, we could not identify specific Ca2+-binding sites in these Ig domains, nor evidence of the association of titin N2A components with F-actin in transfected C2C12 myoblasts or C2C12-derived myotubes. In addition, F-actin cosedimentation assays failed to reveal binding to N2A. We conclude that N2A has a unique architecture that predictably supports its selective recruitment of binding partners in signaling, but that its mechanical role through interaction with F-actin awaits validation.


Subject(s)
Actins , Sarcomeres , Actin Cytoskeleton/metabolism , Actins/genetics , Actins/metabolism , Binding Sites , Connectin/genetics , Connectin/metabolism , Sarcomeres/metabolism
12.
J Mol Biol ; 433(9): 166901, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33647290

ABSTRACT

Striated muscle responds to mechanical overload by rapidly up-regulating the expression of the cardiac ankyrin repeat protein, CARP, which then targets the sarcomere by binding to titin N2A in the I-band region. To date, the role of this interaction in the stress response of muscle remains poorly understood. Here, we characterise the molecular structure of the CARP-receptor site in titin (UN2A) and its binding of CARP. We find that titin UN2A contains a central three-helix bundle fold (ca 45 residues in length) that is joined to N- and C-terminal flanking immunoglobulin domains by long, flexible linkers with partial helical content. CARP binds titin by engaging an α-hairpin in the three-helix fold of UN2A, the C-terminal linker sequence, and the BC loop in Ig81, which jointly form a broad binding interface. Mutagenesis showed that the CARP/N2A association withstands sequence variations in titin N2A and we use this information to evaluate 85 human single nucleotide variants. In addition, actin co-sedimentation, co-transfection in C2C12 cells, proteomics on heart lysates, and the mechanical response of CARP-soaked myofibrils imply that CARP induces the cross-linking of titin and actin myofilaments, thereby increasing myofibril stiffness. We conclude that CARP acts as a regulator of force output in the sarcomere that preserves muscle mechanical performance upon overload stress.


Subject(s)
Actins/chemistry , Actins/metabolism , Connectin/chemistry , Connectin/metabolism , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Male , Mice , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutation , Myofibrils/chemistry , Myofibrils/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Pliability , Protein Binding , Rabbits , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sarcomeres/chemistry , Sarcomeres/metabolism
13.
Life Sci ; 267: 118974, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33385407

ABSTRACT

AIM: We aimed to determine whether the sodium/glucose cotransporter family member SGLT3, a proposed glucose sensor, is expressed in the intestine and/or kidney, and if its expression is altered in mouse models of obesity and in humans before and after weight-loss surgery. MAIN METHODS: We used in-situ hybridization and quantitative PCR to determine whether the Sglt3 isoforms 3a and 3b were expressed in the intestine and kidney of C57, leptin-deficient ob/ob, and diabetic BTBR ob/ob mice. Western blotting and immunohistochemistry were also used to assess SGLT3 protein levels in jejunal biopsies from obese patients before and after weight-loss Roux-en-Y gastric bypass surgery (RYGB), and in lean healthy controls. KEY FINDINGS: Sglt3a/3b mRNA was detected in the small intestine (duodenum, jejunum and ileum), but not in the large intestine or kidneys of mice. Both isoforms were detected in epithelial cells (confirmed using intestinal organoids). Expression of Sglt3a/3b mRNA in duodenum and jejunum was significantly lower in ob/ob and BTBR ob/ob mice than in normal-weight littermates. Jejunal SGLT3 protein levels in aged obese patients before RYGB were lower than in lean individuals, but substantially upregulated 6 months post-RYGB. SIGNIFICANCE: Our study shows that Sglt3a/3b is expressed primarily in epithelial cells of the small intestine in mice. Furthermore, we observed an association between intestinal mRNA Sglt3a/3b expression and obesity in mice, and between jejunal SGLT3 protein levels and obesity in humans. Further studies are required to determine the possible role of SGLT3 in obesity.


Subject(s)
Obesity/metabolism , Sodium-Glucose Transport Proteins/genetics , Adult , Animals , Disease Models, Animal , Down-Regulation , Female , Gastric Bypass , Gene Expression , Humans , Insulin/metabolism , Insulin Resistance , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Jejunum/metabolism , Leptin/deficiency , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/genetics , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Glucose Transport Proteins/biosynthesis , Sodium-Glucose Transport Proteins/metabolism , Transcriptome , Weight Loss
14.
Sci Rep ; 9(1): 15385, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659186

ABSTRACT

The search for biomarkers associated with obesity-related diseases is ongoing, but it is not clear whether plasma and serum can be used interchangeably in this process. Here we used high-throughput screening to analyze 358 proteins and 76 lipids, selected because of their relevance to obesity-associated diseases, in plasma and serum from age- and sex-matched lean and obese humans. Most of the proteins/lipids had similar concentrations in plasma and serum, but a subset showed significant differences. Notably, a key marker of cardiovascular disease PAI-1 showed a difference in concentration between the obese and lean groups only in plasma. Furthermore, some biomarkers showed poor correlations between plasma and serum, including PCSK9, an important regulator of cholesterol homeostasis. Collectively, our results show that the choice of biofluid may impact study outcome when screening for obesity-related biomarkers and we identify several markers where this will be the case.


Subject(s)
Kidney Diseases/blood , Metabolic Syndrome/blood , Obesity/blood , Plasminogen Activator Inhibitor 1/blood , Proprotein Convertase 9/blood , Adult , Biomarkers/blood , Female , Humans , Kidney Diseases/complications , Male , Metabolic Syndrome/complications , Middle Aged , Obesity/complications , Plasminogen Activator Inhibitor 1/standards , Proprotein Convertase 9/standards
15.
JCI Insight ; 4(19)2019 09 12.
Article in English | MEDLINE | ID: mdl-31513548

ABSTRACT

Perturbations in biomechanical stimuli during cardiac development contribute to congenital cardiac defects such as hypoplastic left heart syndrome (HLHS). This study sought to identify stretch-responsive pathways involved in cardiac development. miRNA-Seq identified miR-486 as being increased in cardiomyocytes exposed to cyclic stretch in vitro. The right ventricles (RVs) of patients with HLHS experienced increased stretch and had a trend toward higher miR-486 levels. Sheep RVs dilated from excessive pulmonary blood flow had 60% more miR-486 compared with control RVs. The left ventricles of newborn mice treated with miR-486 mimic were 16.9%-24.6% larger and displayed a 2.48-fold increase in cardiomyocyte proliferation. miR-486 treatment decreased FoxO1 and Smad signaling while increasing the protein levels of Stat1. Stat1 associated with Gata-4 and serum response factor (Srf), 2 key cardiac transcription factors with protein levels that increase in response to miR-486. This is the first report to our knowledge of a stretch-responsive miRNA that increases the growth of the ventricle in vivo.


Subject(s)
Heart Ventricles/growth & development , Hypoplastic Left Heart Syndrome/genetics , MicroRNAs/metabolism , Animals , Animals, Newborn , Biomechanical Phenomena , Cell Proliferation/physiology , Cells, Cultured , Heart Ventricles/metabolism , Humans , Hypoplastic Left Heart Syndrome/pathology , Hypoplastic Left Heart Syndrome/physiopathology , Mechanotransduction, Cellular/physiology , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , STAT1 Transcription Factor/metabolism , Sheep
16.
Diabetologia ; 60(4): 729-739, 2017 04.
Article in English | MEDLINE | ID: mdl-28188334

ABSTRACT

AIMS/HYPOTHESIS: In this study, we aimed to evaluate the therapeutic potential of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an activator of AMP-activated protein kinase, for ameliorating high-fat diet (HFD)-induced pathophysiology in mice. We also aimed to determine whether the beneficial effects of AICAR were dependent on adiponectin. Furthermore, human adipose tissue was used to examine the effect of AICAR ex vivo. METHODS: Six-week-old male C57BL/6J wild-type and Adipoq -/- mice were fed a standard-fat diet (10% fat) or an HFD (60% fat) for 12 weeks and given vehicle or AICAR (500 µg/g) three times/week from weeks 4-12. Diet-induced pathophysiology was examined in mice after 11 weeks by IPGTT and after 12 weeks by flow cytometry and western blotting. Human adipose tissue biopsies from obese (BMI 35-50 kg/m2) individuals were incubated with vehicle or AICAR (1 mmol/l) for 6 h at 37°C, after which inflammation was characterised by ELISA (TNF-α) and flow cytometry. RESULTS: AICAR attenuated adipose inflammation in mice fed an HFD, promoting an M1-to-M2 macrophage phenotype switch, while reducing infiltration of CD8+ T cells. AICAR treatment of mice fed an HFD partially restored glucose tolerance and attenuated hepatic steatosis and kidney disease, as evidenced by reduced albuminuria (p < 0.05), urinary H2O2 (p < 0.05) and renal superoxide levels (p < 0.01) in both wild-type and Adipoq -/- mice. AICAR-mediated protection occurred independently of adiponectin, as similar protection was observed in wild-type and Adipoq -/- mice. In addition, AICAR promoted an M1-to-M2 macrophage phenotype switch and reduced TNF-α production in tissue explants from obese human patients. CONCLUSIONS/INTERPRETATION: AICAR may promote metabolic health and protect against obesity-induced systemic diseases in an adiponectin-independent manner. Furthermore, AICAR reduced inflammation in human adipose tissue explants, suggesting by proof-of-principle that the drug may reduce obesity-induced complications in humans. TRIAL REGISTRATION: ClinicalTrials.gov NCT02322073.


Subject(s)
Adiponectin/metabolism , Diet, High-Fat/adverse effects , Adiponectin/genetics , Animals , Inflammation/immunology , Inflammation/metabolism , Kidney Diseases/immunology , Kidney Diseases/metabolism , Liver Diseases/immunology , Liver Diseases/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Obesity/metabolism
17.
Diabetologia ; 59(3): 582-91, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26631215

ABSTRACT

AIMS/HYPOTHESIS: Tankyrase (TNKS) is a ubiquitously expressed molecular scaffold that is implicated in diverse processes. The catalytic activity of TNKS modifies substrate proteins through poly-ADP-ribosylation (PARsylation) and is responsive to cellular energetic state. Global deficiency of the TNKS protein in mice accelerates glucose utilisation and raises plasma adiponectin levels. The aim of this study was to investigate whether the PARsylation activity of TNKS in adipocytes plays a role in systemic glucose homeostasis. METHODS: To inhibit TNKS-mediated PARsylation, we fed mice with a diet containing the TNKS-specific inhibitor G007-LK. To genetically inactivate TNKS catalysis in adipocytes while preserving its function as a molecular scaffold, we used an adipocyte-selective Cre transgene to delete TNKS exons that encoded the catalytic domain at the C-terminus. Tissue-specific insulin sensitivity in mice was investigated using hyperinsulinaemic-euglycaemic clamps. To model adipose-liver crosstalk ex vivo, we applied adipocyte-conditioned media to hepatocytes and assessed the effect on gluconeogenesis. RESULTS: The TNKS inhibitor G007-LK improved glucose tolerance and insulin sensitivity and promptly increased plasma adiponectin levels. In female mice, but not in male mice, adipocyte-selective genetic inactivation of TNKS catalysis improved hepatic insulin sensitivity and post-transcriptionally increased plasma adiponectin levels. Both pharmacological and genetic TNKS inhibition in female mouse-derived adipocytes induced a change in secreted factors to decrease gluconeogenesis in primary hepatocytes. CONCLUSIONS/INTERPRETATION: Systemic glucose homeostasis is regulated by the PARsylation activity of TNKS in adipocytes. This regulation is mediated in part by adipocyte-secreted factors that modulate hepatic glucose production. Pharmacological TNKS inhibition could potentially be used to improve glucose tolerance.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/enzymology , Glucose/metabolism , Tankyrases/metabolism , Animals , Blood Glucose/drug effects , Carbohydrate Metabolism/drug effects , Female , Male , Mice , Sulfones/pharmacology , Tankyrases/antagonists & inhibitors , Triazoles/pharmacology
18.
Cell Metab ; 22(1): 125-37, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26052006

ABSTRACT

The role of inflammation in obesity-related pathologies is well established. We investigated the therapeutic potential of LipoxinA4 (LXA4:5(S),6(R),15(S)-trihydroxy-7E,9E,11Z,13E,-eicosatetraenoic acid) and a synthetic 15(R)-Benzo-LXA4-analog as interventions in a 3-month high-fat diet (HFD; 60% fat)-induced obesity model. Obesity caused distinct pathologies, including impaired glucose tolerance, adipose inflammation, fatty liver, and chronic kidney disease (CKD). Lipoxins (LXs) attenuated obesity-induced CKD, reducing glomerular expansion, mesangial matrix, and urinary H2O2. Furthermore, LXA4 reduced liver weight, serum alanine-aminotransferase, and hepatic triglycerides. LXA4 decreased obesity-induced adipose inflammation, attenuating TNF-α and CD11c(+) M1-macrophages (MΦs), while restoring CD206(+) M2-MΦs and increasing Annexin-A1. LXs did not affect renal or hepatic MΦs, suggesting protection occurred via attenuation of adipose inflammation. LXs restored adipose expression of autophagy markers LC3-II and p62. LX-mediated protection was demonstrable in adiponectin(-/-) mice, suggesting that the mechanism was adiponectin independent. In conclusion, LXs protect against obesity-induced systemic disease, and these data support a novel therapeutic paradigm for treating obesity and associated pathologies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Kidney Diseases/complications , Lipoxins/therapeutic use , Liver Diseases/complications , Obesity/complications , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Autophagy/drug effects , Inflammation/immunology , Inflammation/pathology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Kidney Diseases/drug therapy , Kidney Diseases/immunology , Kidney Diseases/pathology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Diseases/drug therapy , Liver Diseases/immunology , Liver Diseases/pathology , Mice, Inbred C57BL , Obesity/immunology
19.
Curr Opin Pharmacol ; 13(4): 618-24, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23751262

ABSTRACT

Obesity-induced inflammation is associated with numerous pathologies and is an independent risk factor of chronic kidney disease (CKD). The prevalence of CKD is escalating and current therapeutic strategies are seriously lacking in efficacy, and immunomodulation has been suggested as a potential new therapeutic approach. Indeed, specialized pro-resolving mediators (SPMs), such as lipoxins (LXs), resolvins and protectins, have demonstrated protection in adipose inflammation, restoring insulin sensitivity and adiponectin production, while modulating leukocyte infiltration and promoting resolution in visceral adipose tissue. Furthermore, SPMs display direct renoprotective effect. Thus we review current evidence of immunomodulation as a potential strategy to subvert obesity-related CKD.


Subject(s)
Obesity/immunology , Renal Insufficiency, Chronic/immunology , Adipose Tissue/immunology , Animals , Humans , Immunomodulation , Inflammation
20.
Biochim Biophys Acta ; 1833(8): 1969-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23567938

ABSTRACT

TGF-ß1 is a prototypic profibrotic cytokine and major driver of fibrosis in the kidney and other organs. Induced in high glucose-1 (IHG-1) is a mitochondrial protein which we have recently reported to be associated with renal disease. IHG-1 amplifies responses to TGF-ß1 and regulates mitochondrial biogenesis by stabilising the transcriptional co-activator peroxisome proliferator-activated receptor gamma coactivator-1-alpha. Here we report that the mitochondrial localisation of IHG-1 is pivotal in the amplification of TGF-ß1 signalling. We demonstrate that IHG-1 expression is associated with repression of the endogenous TGF-ß1 inhibitor Smad7. Intriguingly, expression of a non-mitochondrial deletion mutant of IHG-1 (Δmts-IHG-1) repressed TGF-ß1 fibrotic signalling in renal epithelial cells. In cells expressing Δmts-IHG-1 fibrotic responses including CCN2/connective tissue growth factor, fibronectin and jagged-1 expression were reduced following stimulation with TGF-ß1. Δmts-IHG-1 modulation of TGF-ß1 signalling was associated with increased Smad7 protein expression. Δmts-IHG-1 modulated TGF-ß1 activity by increasing Smad7 protein expression as it failed to inhibit TGF-ß1 transcriptional responses when endogenous Smad7 expression was knocked down. These data indicate that mitochondria modulate TGF-ß1 signal transduction and that IHG-1 is a key player in this modulation.


Subject(s)
Fibrosis/metabolism , Mitochondria/genetics , Proteins/metabolism , Smad7 Protein/biosynthesis , Transforming Growth Factor beta1/metabolism , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Molecular Sequence Data , Phosphorylation , Proteins/genetics , Serrate-Jagged Proteins , Signal Transduction , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...