Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 181(6): 1246-1262.e22, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32442405

ABSTRACT

There is considerable inter-individual variability in susceptibility to weight gain despite an equally obesogenic environment in large parts of the world. Whereas many studies have focused on identifying the genetic susceptibility to obesity, we performed a GWAS on metabolically healthy thin individuals (lowest 6th percentile of the population-wide BMI spectrum) in a uniquely phenotyped Estonian cohort. We discovered anaplastic lymphoma kinase (ALK) as a candidate thinness gene. In Drosophila, RNAi mediated knockdown of Alk led to decreased triglyceride levels. In mice, genetic deletion of Alk resulted in thin animals with marked resistance to diet- and leptin-mutation-induced obesity. Mechanistically, we found that ALK expression in hypothalamic neurons controls energy expenditure via sympathetic control of adipose tissue lipolysis. Our genetic and mechanistic experiments identify ALK as a thinness gene, which is involved in the resistance to weight gain.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Thinness/genetics , Adipose Tissue/metabolism , Adult , Animals , Cell Line , Cohort Studies , Drosophila/genetics , Estonia , Female , Humans , Leptin/genetics , Lipolysis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , RNA Interference/physiology , Young Adult
2.
Cell ; 177(6): 1536-1552.e23, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150623

ABSTRACT

Ectopic lipid deposition and altered mitochondrial dynamics contribute to the development of obesity and insulin resistance. However, the mechanistic link between these processes remained unclear. Here we demonstrate that the C16:0 sphingolipid synthesizing ceramide synthases, CerS5 and CerS6, affect distinct sphingolipid pools and that abrogation of CerS6 but not of CerS5 protects from obesity and insulin resistance. We identify proteins that specifically interact with C16:0 sphingolipids derived from CerS5 or CerS6. Here, only CerS6-derived C16:0 sphingolipids bind the mitochondrial fission factor (Mff). CerS6 and Mff deficiency protect from fatty acid-induced mitochondrial fragmentation in vitro, and the two proteins genetically interact in vivo in obesity-induced mitochondrial fragmentation and development of insulin resistance. Our experiments reveal an unprecedented specificity of sphingolipid signaling depending on specific synthesizing enzymes, provide a mechanistic link between hepatic lipid deposition and mitochondrial fragmentation in obesity, and define the CerS6-derived sphingolipid/Mff interaction as a therapeutic target for metabolic diseases.


Subject(s)
Membrane Proteins/metabolism , Obesity/metabolism , Sphingolipids/metabolism , Sphingosine N-Acyltransferase/metabolism , Animals , Apoptosis , Cell Line , HeLa Cells , Humans , Insulin Resistance/physiology , Liver/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Obesity/physiopathology , Sphingolipids/physiology , Sphingosine N-Acyltransferase/physiology
3.
Cell ; 165(4): 882-95, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133169

ABSTRACT

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Subject(s)
Brain/metabolism , Diet, High-Fat , Glucose/metabolism , Obesity/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/metabolism , Cognition , Endothelial Cells/metabolism , Fatty Acids/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Myeloid Cells/metabolism
4.
Cell ; 158(4): 874-888, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25126791

ABSTRACT

Stem cells are highly abundant during early development but become a rare population in most adult organs. The molecular mechanisms causing stem cells to exit proliferation at a specific time are not well understood. Here, we show that changes in energy metabolism induced by the steroid hormone ecdysone and the Mediator initiate an irreversible cascade of events leading to cell-cycle exit in Drosophila neural stem cells. We show that the timely induction of oxidative phosphorylation and the mitochondrial respiratory chain are required in neuroblasts to uncouple the cell cycle from cell growth. This results in a progressive reduction in neuroblast cell size and ultimately in terminal differentiation. Brain tumor mutant neuroblasts fail to undergo this shrinkage process and continue to proliferate until adulthood. Our findings show that cell size control can be modified by systemic hormonal signaling and reveal a unique connection between metabolism and proliferation in stem cells.


Subject(s)
Cell Proliferation , Drosophila melanogaster/cytology , Ecdysone/metabolism , Neural Stem Cells/cytology , Animals , Cell Size , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Energy Metabolism , Genome, Insect , Mediator Complex/metabolism , Neural Stem Cells/metabolism
5.
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
8.
Genes Dev ; 31(20): 2099-2112, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29118048

ABSTRACT

Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D -driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D -driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Receptor Activator of Nuclear Factor-kappa B/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cell Respiration , Cells, Cultured , Energy Metabolism , Female , Gonadal Steroid Hormones/physiology , Homeostasis , Humans , Lung/metabolism , Lung Neoplasms/drug therapy , Male , Mice , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptor Activator of Nuclear Factor-kappa B/antagonists & inhibitors , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Respiratory Mucosa/metabolism
9.
Nature ; 563(7732): 564-568, 2018 11.
Article in English | MEDLINE | ID: mdl-30405245

ABSTRACT

Genetic regulators and environmental stimuli modulate T cell activation in autoimmunity and cancer. The enzyme co-factor tetrahydrobiopterin (BH4) is involved in the production of monoamine neurotransmitters, the generation of nitric oxide, and pain1,2. Here we uncover a link between these processes, identifying a fundamental role for BH4 in T cell biology. We find that genetic inactivation of GTP cyclohydrolase 1 (GCH1, the rate-limiting enzyme in the synthesis of BH4) and inhibition of sepiapterin reductase (the terminal enzyme in the synthetic pathway for BH4) severely impair the proliferation of mature mouse and human T cells. BH4 production in activated T cells is linked to alterations in iron metabolism and mitochondrial bioenergetics. In vivo blockade of BH4 synthesis abrogates T-cell-mediated autoimmunity and allergic inflammation, and enhancing BH4 levels through GCH1 overexpression augments responses by CD4- and CD8-expressing T cells, increasing their antitumour activity in vivo. Administration of BH4 to mice markedly reduces tumour growth and expands the population of intratumoral effector T cells. Kynurenine-a tryptophan metabolite that blocks antitumour immunity-inhibits T cell proliferation in a manner that can be rescued by BH4. Finally, we report the development of a potent SPR antagonist for possible clinical use. Our data uncover GCH1, SPR and their downstream metabolite BH4 as critical regulators of T cell biology that can be readily manipulated to either block autoimmunity or enhance anticancer immunity.


Subject(s)
Autoimmune Diseases/immunology , Biopterins/analogs & derivatives , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Administration, Oral , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Biopterins/biosynthesis , Biopterins/metabolism , Biopterins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Coenzymes/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Humans , Hypersensitivity/immunology , Iron/metabolism , Kynurenine/metabolism , Kynurenine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
J Allergy Clin Immunol ; 145(1): 283-300.e8, 2020 01.
Article in English | MEDLINE | ID: mdl-31401286

ABSTRACT

BACKGROUND: Perturbation of epidermal barrier formation will profoundly compromise overall skin function, leading to a dry and scaly, ichthyosis-like skin phenotype that is the hallmark of a broad range of skin diseases, including ichthyosis, atopic dermatitis, and a multitude of clinical eczema variants. An overarching molecular mechanism that orchestrates the multitude of factors controlling epidermal barrier formation and homeostasis remains to be elucidated. OBJECTIVE: Here we highlight a specific role of mammalian target of rapamycin complex 2 (mTORC2) signaling in epidermal barrier formation. METHODS: Epidermal mTORC2 signaling was specifically disrupted by deleting rapamycin-insensitive companion of target of rapamycin (Rictor), encoding an essential subunit of mTORC2 in mouse epidermis (epidermis-specific homozygous Rictor deletion [RicEKO] mice). Epidermal structure and barrier function were investigated through a combination of gene expression, biochemical, morphological and functional analysis in RicEKO and control mice. RESULTS: RicEKO newborns displayed an ichthyosis-like phenotype characterized by dysregulated epidermal de novo lipid synthesis, altered lipid lamellae structure, and aberrant filaggrin (FLG) processing. Despite a compensatory transcriptional epidermal repair response, the protective epidermal function was impaired in RicEKO mice, as revealed by increased transepidermal water loss, enhanced corneocyte fragility, decreased dendritic epidermal T cells, and an exaggerated percutaneous immune response. Restoration of Akt-Ser473 phosphorylation in mTORC2-deficient keratinocytes through expression of constitutive Akt rescued FLG processing. CONCLUSION: Our findings reveal a critical metabolic signaling relay of barrier formation in which epidermal mTORC2 activity controls FLG processing and de novo epidermal lipid synthesis during cornification. Our findings provide novel mechanistic insights into epidermal barrier formation and could open up new therapeutic opportunities to restore defective epidermal barrier conditions.


Subject(s)
Epidermis , Intermediate Filament Proteins , Lipids , Protein Processing, Post-Translational/immunology , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction/immunology , Animals , Epidermis/immunology , Epidermis/metabolism , Filaggrin Proteins , Ichthyosis/genetics , Ichthyosis/immunology , Ichthyosis/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/immunology , Intermediate Filament Proteins/metabolism , Lipids/biosynthesis , Lipids/genetics , Lipids/immunology , Mice , Mice, Knockout , Protein Processing, Post-Translational/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/immunology , Signal Transduction/genetics
11.
Gut ; 68(3): 533-546, 2019 03.
Article in English | MEDLINE | ID: mdl-29374630

ABSTRACT

OBJECTIVE: Liver injury impacts hepatic inflammation in part via Toll-like receptor (TLR) signalling. Triggering receptor expressed on myeloid cells 2 (TREM-2) modulates TLR4-mediated inflammation in bone marrow (BM)-derived macrophages but its function in liver injury is unknown. Here we hypothesised that the anti-inflammatory effects of TREM-2 on TLR signalling may limit hepatic injury. DESIGN: TREM-2 expression was analysed in livers of humans with various forms of liver injury compared with control individuals. Acute and chronic liver injury models were performed in wild type and Trem-2-/- mice. Primary liver cells from both genotypes of mice were isolated for in vitro experiments. RESULTS: TREM-2 was expressed on non-parenchymal hepatic cells and induced during liver injury in mice and man. Mice lacking TREM-2 exhibited heightened liver damage and inflammation during acute and repetitive carbon tetrachloride and acetaminophen (APAP) intoxication, the latter of which TREM-2 deficiency was remarkably associated with worsened survival. Liver damage in Trem-2-/- mice following chronic injury and APAP challenge was associated with elevated hepatic lipid peroxidation and macrophage content. BM transplantation experiments and cellular reactive oxygen species assays revealed effects of TREM-2 in the context of chronic injury depended on both immune and resident TREM-2 expression. Consistent with effects of TREM-2 on inflammation-associated injury, primary hepatic macrophages and hepatic stellate cells lacking TREM-2 exhibited augmented TLR4-driven proinflammatory responses. CONCLUSION: Our data indicate that by acting as a natural brake on inflammation during hepatocellular injury, TREM-2 is a critical regulator of diverse types of hepatotoxic injury.


Subject(s)
Liver Cirrhosis/metabolism , Liver/metabolism , Membrane Glycoproteins/physiology , Receptors, Immunologic/physiology , Acetaminophen , Aged , Animals , Carbon Tetrachloride , Case-Control Studies , Female , Hematopoietic Stem Cells/metabolism , Hepatocytes/metabolism , Humans , Inflammation Mediators/metabolism , Kupffer Cells/metabolism , Lipid Peroxidation/physiology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Knockout , Middle Aged , Reactive Oxygen Species/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Toll-Like Receptor 4/physiology , Up-Regulation/physiology
12.
Cell Rep ; 43(6): 114343, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38865247

ABSTRACT

Activation of prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus (ARC) promotes high-fat-diet (HFD)-induced hyperphagia. In turn, PNOCARC neurons can inhibit the anorexic response of proopiomelanocortin (POMC) neurons. Here, we validate the necessity of PNOCARC activity for HFD-induced inhibition of POMC neurons in mice and find that PNOCARC-neuron-dependent inhibition of POMC neurons is mediated by gamma-aminobutyric acid (GABA) release. When monitoring individual PNOCARC neuron activity via Ca2+ imaging, we find a subpopulation of PNOCARC neurons that is inhibited upon gastrointestinal calorie sensing and disinhibited upon HFD feeding. Combining retrograde rabies tracing and circuit mapping, we find that PNOC neurons from the bed nucleus of the stria terminalis (PNOCBNST) provide inhibitory input to PNOCARC neurons, and this inhibitory input is blunted upon HFD feeding. This work sheds light on how an increase in caloric content of the diet can rewire a neuronal circuit, paving the way to overconsumption and obesity development.


Subject(s)
Diet, High-Fat , Hyperphagia , Septal Nuclei , Animals , Hyperphagia/metabolism , Mice , Septal Nuclei/metabolism , Neurons/metabolism , Male , gamma-Aminobutyric Acid/metabolism , Pro-Opiomelanocortin/metabolism , GABAergic Neurons/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Mice, Inbred C57BL , Protein Precursors , Receptors, Opioid
13.
Endocr Rev ; 43(2): 314-328, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34490882

ABSTRACT

The central nervous system (CNS) receives information from afferent neurons, circulating hormones, and absorbed nutrients and integrates this information to orchestrate the actions of the neuroendocrine and autonomic nervous systems in maintaining systemic metabolic homeostasis. Particularly the arcuate nucleus of the hypothalamus (ARC) is of pivotal importance for primary sensing of adiposity signals, such as leptin and insulin, and circulating nutrients, such as glucose. Importantly, energy state-sensing neurons in the ARC not only regulate feeding but at the same time control multiple physiological functions, such as glucose homeostasis, blood pressure, and innate immune responses. These findings have defined them as master regulators, which adapt integrative physiology to the energy state of the organism. The disruption of this fine-tuned control leads to an imbalance between energy intake and expenditure as well as deregulation of peripheral metabolism. Improving our understanding of the cellular, molecular, and functional basis of this regulatory principle in the CNS could set the stage for developing novel therapeutic strategies for the treatment of obesity and metabolic syndrome. In this review, we summarize novel insights with a particular emphasis on ARC neurocircuitries regulating food intake and glucose homeostasis and sensing factors that inform the brain of the organismal energy status.


Subject(s)
Arcuate Nucleus of Hypothalamus , Diabetes Mellitus , Arcuate Nucleus of Hypothalamus/metabolism , Diabetes Mellitus/metabolism , Energy Metabolism/physiology , Glucose/metabolism , Humans , Hypothalamus/metabolism , Leptin/metabolism , Obesity/metabolism
14.
Front Cell Dev Biol ; 10: 979251, 2022.
Article in English | MEDLINE | ID: mdl-36200038

ABSTRACT

In humans, various dietary and social factors led to the development of increased brain sizes alongside large adipose tissue stores. Complex reciprocal signaling mechanisms allow for a fine-tuned interaction between the two organs to regulate energy homeostasis of the organism. As an endocrine organ, adipose tissue secretes various hormones, cytokines, and metabolites that signal energy availability to the central nervous system (CNS). Vice versa, the CNS is a critical regulator of adipose tissue function through neural networks that integrate information from the periphery and regulate sympathetic nerve outflow. This review discusses the various reciprocal signaling mechanisms in the CNS and adipose tissue to maintain organismal energy homeostasis. We are focusing on the integration of afferent signals from the periphery in neuronal populations of the mediobasal hypothalamus as well as the efferent signals from the CNS to adipose tissue and its implications for adipose tissue function. Furthermore, we are discussing central mechanisms that fine-tune the immune system in adipose tissue depots and contribute to organ homeostasis. Elucidating this complex signaling network that integrates peripheral signals to generate physiological outputs to maintain the optimal energy balance of the organism is crucial for understanding the pathophysiology of obesity and metabolic diseases such as type 2 diabetes.

15.
Mol Metab ; 66: 101626, 2022 12.
Article in English | MEDLINE | ID: mdl-36356831

ABSTRACT

OBJECTIVE: Nonalcoholic fatty liver disease (NAFLD) ranges from steatosis to nonalcoholic steatohepatitis (NASH), which often progresses to hepatocellular carcinoma (HCC) through a largely undefined mechanism. NASH and HCC depend on inflammatory signaling, whose master regulator is the NFκB transcription factor family, activated by canonical and non-canonical pathways. METHODS: Here, we investigated non-canonical NFκB-inducing kinase (NIK/MAP3K14) in metabolic NASH, NASH to HCC transition, and DEN-induced HCC. To this end, we performed dietary and chemical interventions in mice that were analyzed via single nucleus sequencing, gene expression and histochemical methods. Ultimately, we verified our mouse results in human patient samples. RESULTS: We revealed that hepatocyte-specific NIK deficiency (NIKLKO) ameliorated metabolic NASH complications and reduced hepatocarcinogenesis, independent of its role in the NFκB pathway. Instead, hepatic NIK attenuated hepatoprotective JAK2/STAT5 signaling that is a prerequisite for NASH and NASH to HCC progression in mice and humans. CONCLUSIONS: Our data suggest NIK-mediated inhibitory JAK2 phosphorylation at serine 633 that might be amenable for future therapeutic interventions in patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Carcinoma, Hepatocellular/metabolism , Hepatocytes/metabolism , Janus Kinase 2/metabolism , Liver Neoplasms/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , STAT5 Transcription Factor/metabolism , NF-kappaB-Inducing Kinase
16.
Cell Metab ; 33(12): 2398-2414.e9, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34715039

ABSTRACT

Wound healing is a coordinated process that initially relies on pro-inflammatory macrophages, followed by a pro-resolution function of these cells. Changes in cellular metabolism likely dictate these distinct activities, but the nature of these changes has been unclear. Here, we profiled early- versus late-stage skin wound macrophages in mice at both the transcriptional and functional levels. We found that glycolytic metabolism in the early phase is not sufficient to ensure productive repair. Instead, by combining conditional disruption of the electron transport chain with deletion of mitochondrial aspartyl-tRNA synthetase, followed by single-cell sequencing analysis, we found that a subpopulation of early-stage wound macrophages are marked by mitochondrial ROS (mtROS) production and HIF1α stabilization, which ultimately drives a pro-angiogenic program essential for timely healing. In contrast, late-phase, pro-resolving wound macrophages are marked by IL-4Rα-mediated mitochondrial respiration and mitohormesis. Collectively, we identify changes in mitochondrial metabolism as a critical control mechanism for macrophage effector functions during wound healing.


Subject(s)
Macrophages , Wound Healing , Animals , Macrophages/metabolism , Mice , Mitochondria/metabolism
17.
Nat Metab ; 3(12): 1662-1679, 2021 12.
Article in English | MEDLINE | ID: mdl-34931084

ABSTRACT

Insulin acts on neurons and glial cells to regulate systemic glucose metabolism and feeding. However, the mechanisms of insulin access in discrete brain regions are incompletely defined. Here we show that insulin receptors in tanycytes, but not in brain endothelial cells, are required to regulate insulin access to the hypothalamic arcuate nucleus. Mice lacking insulin receptors in tanycytes (IR∆Tan mice) exhibit systemic insulin resistance, while displaying normal food intake and energy expenditure. Tanycytic insulin receptors are also necessary for the orexigenic effects of ghrelin, but not for the anorexic effects of leptin. IR∆Tan mice exhibit increased agouti-related peptide (AgRP) neuronal activity, while displaying blunted AgRP neuronal adaptations to feeding-related stimuli. Lastly, a highly palatable food decreases tanycytic and arcuate nucleus insulin signalling to levels comparable to those seen in IR∆Tan mice. These changes are rooted in modifications of cellular stress responses and of mitochondrial protein quality control in tanycytes. Conclusively, we reveal a critical role of tanycyte insulin receptors in gating feeding-state-dependent regulation of AgRP neurons and systemic insulin sensitivity, and show that insulin resistance in tanycytes contributes to the pleiotropic manifestations of obesity-associated insulin resistance.


Subject(s)
Agouti-Related Protein/metabolism , Ependymoglial Cells/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Neurons/metabolism , Signal Transduction , Agouti-Related Protein/chemistry , Animals , Biomarkers , Blood-Brain Barrier/metabolism , Calcium , Energy Metabolism , Fluorescent Antibody Technique , Ghrelin/metabolism , Glucose/metabolism , Insulin Resistance , Mice , Mice, Knockout , Mitochondria/metabolism , Models, Biological , Peptide Fragments/metabolism , Receptor, Insulin/metabolism
18.
Diabetes ; 70(9): 2042-2057, 2021 09.
Article in English | MEDLINE | ID: mdl-33627323

ABSTRACT

Obesity-induced white adipose tissue (WAT) hypertrophy is associated with elevated adipose tissue macrophage (ATM) content. Overexpression of the triggering receptor expressed on myeloid cells 2 (TREM2) reportedly increases adiposity, worsening health. Paradoxically, using insulin resistance, elevated fat mass, and hypercholesterolemia as hallmarks of unhealthy obesity, a recent report demonstrated that ATM-expressed TREM2 promoted health. Here, we identified that in mice, TREM2 deficiency aggravated diet-induced insulin resistance and hepatic steatosis independently of fat and cholesterol levels. Metabolomics linked TREM2 deficiency with elevated obesity-instigated serum ceramides that correlated with impaired insulin sensitivity. Remarkably, while inhibiting ceramide synthesis exerted no influences on TREM2-dependent ATM remodeling, inflammation, or lipid load, it restored insulin tolerance, reversing adipose hypertrophy and secondary hepatic steatosis of TREM2-deficient animals. Bone marrow transplantation experiments revealed unremarkable influences of immune cell-expressed TREM2 on health, instead demonstrating that WAT-intrinsic mechanisms impinging on sphingolipid metabolism dominate in the systemic protective effects of TREM2 on metabolic health.


Subject(s)
Adipose Tissue/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Obesity/metabolism , Receptors, Immunologic/metabolism , Animals , Diet, High-Fat , Inflammation/metabolism , Insulin Resistance/physiology , Lipid Metabolism/physiology , Mice , Up-Regulation
19.
Mol Cancer ; 9: 200, 2010 Jul 28.
Article in English | MEDLINE | ID: mdl-20667089

ABSTRACT

BACKGROUND: Heme Oxygenase-1 (HO-1) is expressed in many cancers and promotes growth and survival of neoplastic cells. Recently, HO-1 has been implicated in tumor cell invasion and metastasis. However, the molecular mechanisms underlying these biologic effects of HO-1 remain largely unknown. To identify a common mechanism of action of HO-1 in cancer, we determined the global effect of HO-1 on the transcriptome of multiple tumor entities and identified a universal HO-1-associated gene expression signature. RESULTS: Genome-wide expression profiling of Heme Oxygenase-1 expressing versus HO-1 silenced BeWo choriocarcinoma cells as well as a comparative meta-profiling of the preexisting expression database of 190 human tumors of 14 independent cancer types led to the identification of 14 genes, the expression of which correlated strongly and universally with that of HO-1 (P = 0.00002). These genes included regulators of cell plasticity and extracellular matrix (ECM) remodeling (MMP2, ADAM8, TGFB1, BGN, COL21A1, PXDN), signaling (CRIP2, MICB), amino acid transport and glycosylation (SLC7A1 and ST3GAL2), estrogen and phospholipid biosynthesis (AGPAT2 and HSD17B1), protein stabilization (IFI30), and phosphorylation (ALPPL2). We selected PXDN, an adhesion molecule involved in ECM formation, for further analysis and functional characterization. Immunofluorescence and Western blotting confirmed the positive correlation of expression of PXDN and HO-1 in BeWo cancer cells as well as co-localization of these two proteins in invasive extravillous trophoblast cells. Modulation of HO-1 expression in both loss-of and gain-of function cell models (BeWo and 607B melanoma cells, respectively) demonstrated a direct relationship of HO-1 expression with cell adhesion to Fibronectin and Laminin coated wells. The adhesion-promoting effects of HO-1 were dependent on PXDN expression, as loss of PXDN in HO-1 expressing BeWo and 607B cells led to reduced cell attachment to Laminin and Fibronectin coated wells. CONCLUSIONS: Collectively, our results show that HO-1 expression determines a distinct 'molecular signature' in cancer cells, which is enriched in genes associated with tumorigenesis. The protein network downstream of HO-1 modulates adhesion, signaling, transport, and other critical cellular functions of neoplastic cells and thus promotes tumor cell growth and dissemination.


Subject(s)
Cell Adhesion , Gene Expression Profiling , Heme Oxygenase (Decyclizing)/metabolism , Neoplasms/genetics , Base Sequence , DNA Primers , Gene Knockdown Techniques , Heme Oxygenase (Decyclizing)/genetics , Humans , Neoplasms/enzymology , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
20.
Neuron ; 106(6): 1009-1025.e10, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32302532

ABSTRACT

Calorie-rich diets induce hyperphagia and promote obesity, although the underlying mechanisms remain poorly defined. We find that short-term high-fat-diet (HFD) feeding of mice activates prepronociceptin (PNOC)-expressing neurons in the arcuate nucleus of the hypothalamus (ARC). PNOCARC neurons represent a previously unrecognized GABAergic population of ARC neurons distinct from well-defined feeding regulatory AgRP or POMC neurons. PNOCARC neurons arborize densely in the ARC and provide inhibitory synaptic input to nearby anorexigenic POMC neurons. Optogenetic activation of PNOCARC neurons in the ARC and their projections to the bed nucleus of the stria terminalis promotes feeding. Selective ablation of these cells promotes the activation of POMC neurons upon HFD exposure, reduces feeding, and protects from obesity, but it does not affect food intake or body weight under normal chow consumption. We characterize PNOCARC neurons as a novel ARC neuron population activated upon palatable food consumption to promote hyperphagia.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Diet, High-Fat , Feeding Behavior/physiology , GABAergic Neurons/physiology , Hyperphagia , Obesity , Weight Gain/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , GABAergic Neurons/metabolism , Mice , Neural Inhibition/physiology , Neurons/metabolism , Neurons/physiology , Optogenetics , Pro-Opiomelanocortin/metabolism , Protein Precursors/metabolism , Receptors, Opioid/metabolism , Septal Nuclei/physiology
SELECTION OF CITATIONS
SEARCH DETAIL