Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49.338
Filter
Add more filters

Publication year range
1.
Cell ; 187(10): 2359-2374.e18, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38653240

ABSTRACT

Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.


Subject(s)
Adipose Tissue, Brown , Amino Acids, Branched-Chain , Insulin Resistance , Mitochondria , Nitrogen , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Amino Acids, Branched-Chain/metabolism , Mice , Nitrogen/metabolism , Mitochondria/metabolism , Male , Humans , Energy Metabolism , Mice, Inbred C57BL , Oxidative Stress , Insulin/metabolism , Diet, High-Fat , Adipocytes, Brown/metabolism , Signal Transduction
2.
Cell ; 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39326417

ABSTRACT

We report the 1-year results from one patient as the preliminary analysis of a first-in-human phase I clinical trial (ChiCTR2300072200) assessing the feasibility of autologous transplantation of chemically induced pluripotent stem-cell-derived islets (CiPSC islets) beneath the abdominal anterior rectus sheath for type 1 diabetes treatment. The patient achieved sustained insulin independence starting 75 days post-transplantation. The patient's time-in-target glycemic range increased from a baseline value of 43.18% to 96.21% by month 4 post-transplantation, accompanied by a decrease in glycated hemoglobin, an indicator of long-term systemic glucose levels at a non-diabetic level. Thereafter, the patient presented a state of stable glycemic control, with time-in-target glycemic range at >98% and glycated hemoglobin at around 5%. At 1 year, the clinical data met all study endpoints with no indication of transplant-related abnormalities. Promising results from this patient suggest that further clinical studies assessing CiPSC-islet transplantation in type 1 diabetes are warranted.

3.
Annu Rev Biochem ; 92: 247-272, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37001136

ABSTRACT

The insulin receptor (IR) is a type II receptor tyrosine kinase that plays essential roles in metabolism, growth, and proliferation. Dysregulation of IR signaling is linked to many human diseases, such as diabetes and cancers. The resolution revolution in cryo-electron microscopy has led to the determination of several structures of IR with different numbers of bound insulin molecules in recent years, which have tremendously improved our understanding of how IR is activated by insulin. Here, we review the insulin-induced activation mechanism of IR, including (a) the detailed binding modes and functions of insulin at site 1 and site 2 and (b) the insulin-induced structural transitions that are required for IR activation. We highlight several other key aspects of the activation and regulation of IR signaling and discuss the remaining gaps in our understanding of the IR activation mechanism and potential avenues of future research.


Subject(s)
Insulin , Receptor, Insulin , Humans , Receptor, Insulin/genetics , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Cryoelectron Microscopy , Insulin/chemistry , Insulin/metabolism , Signal Transduction , Receptor Protein-Tyrosine Kinases/metabolism , Phosphorylation
4.
Cell ; 186(26): 5812-5825.e21, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38056462

ABSTRACT

Acyl-coenzyme A (acyl-CoA) species are cofactors for numerous enzymes that acylate thousands of proteins. Here, we describe an enzyme that uses S-nitroso-CoA (SNO-CoA) as its cofactor to S-nitrosylate multiple proteins (SNO-CoA-assisted nitrosylase, SCAN). Separate domains in SCAN mediate SNO-CoA and substrate binding, allowing SCAN to selectively catalyze SNO transfer from SNO-CoA to SCAN to multiple protein targets, including the insulin receptor (INSR) and insulin receptor substrate 1 (IRS1). Insulin-stimulated S-nitrosylation of INSR/IRS1 by SCAN reduces insulin signaling physiologically, whereas increased SCAN activity in obesity causes INSR/IRS1 hypernitrosylation and insulin resistance. SCAN-deficient mice are thus protected from diabetes. In human skeletal muscle and adipose tissue, SCAN expression increases with body mass index and correlates with INSR S-nitrosylation. S-nitrosylation by SCAN/SNO-CoA thus defines a new enzyme class, a unique mode of receptor tyrosine kinase regulation, and a revised paradigm for NO function in physiology and disease.


Subject(s)
Insulin , Oxidoreductases Acting on CH-CH Group Donors , Signal Transduction , Animals , Humans , Mice , Acyl Coenzyme A/metabolism , Adipose Tissue/metabolism , Insulin Resistance , Nitric Oxide/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism
5.
Cell ; 184(10): 2537-2564, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33989548

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the leading chronic liver disease worldwide. Its more advanced subtype, nonalcoholic steatohepatitis (NASH), connotes progressive liver injury that can lead to cirrhosis and hepatocellular carcinoma. Here we provide an in-depth discussion of the underlying pathogenetic mechanisms that lead to progressive liver injury, including the metabolic origins of NAFLD, the effect of NAFLD on hepatic glucose and lipid metabolism, bile acid toxicity, macrophage dysfunction, and hepatic stellate cell activation, and consider the role of genetic, epigenetic, and environmental factors that promote fibrosis progression and risk of hepatocellular carcinoma in NASH.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Carcinoma, Hepatocellular/pathology , Humans , Liver/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology
6.
Cell ; 184(3): 840-843, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545037

ABSTRACT

We have recently identified a novel lymphocyte that is a dual expresser (DE) of TCRαß and BCR. DEs in T1D patients are predominated by a public BCR clonotype (clone-x) that encodes a potent autoantigen that cross-activates insulin-reactive T cells. Betts and colleagues were able to detect DEs but alleged to not detect high DE frequency, clone-x, or similar clones in T1D patients. Unfortunately, the authors did not follow our methods and when they did, their flow cytometric data at two sites were conflicting. Moreover, contrary to their claim, we identified clones similar to clone-x in their data along with clones bearing the core motif (DTAMVYYFDYW). Additionally, their report of no increased usage of clone-x VH/DH genes by bulk B cells confirms rather than challenges our results. Finally, the authors failed to provide data verifying purity of their sorted DEs, making it difficult to draw reliable conclusion of their repertoire analysis. This Matters Arising Response paper addresses the Japp et al. (2021) Matters Arising paper, published concurrently in Cell.


Subject(s)
Diabetes Mellitus, Type 1 , B-Lymphocytes , Clone Cells , Humans , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes
7.
Cell ; 181(5): 1112-1130.e16, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32470399

ABSTRACT

Acute physical activity leads to several changes in metabolic, cardiovascular, and immune pathways. Although studies have examined selected changes in these pathways, the system-wide molecular response to an acute bout of exercise has not been fully characterized. We performed longitudinal multi-omic profiling of plasma and peripheral blood mononuclear cells including metabolome, lipidome, immunome, proteome, and transcriptome from 36 well-characterized volunteers, before and after a controlled bout of symptom-limited exercise. Time-series analysis revealed thousands of molecular changes and an orchestrated choreography of biological processes involving energy metabolism, oxidative stress, inflammation, tissue repair, and growth factor response, as well as regulatory pathways. Most of these processes were dampened and some were reversed in insulin-resistant participants. Finally, we discovered biological pathways involved in cardiopulmonary exercise response and developed prediction models revealing potential resting blood-based biomarkers of peak oxygen consumption.


Subject(s)
Energy Metabolism/physiology , Exercise/physiology , Aged , Biomarkers/metabolism , Female , Humans , Insulin/metabolism , Insulin Resistance , Leukocytes, Mononuclear/metabolism , Longitudinal Studies , Male , Metabolome , Middle Aged , Oxygen/metabolism , Oxygen Consumption , Proteome , Transcriptome
8.
Cell ; 177(4): 896-909.e20, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31030999

ABSTRACT

In mammals, endogenous circadian clocks sense and respond to daily feeding and lighting cues, adjusting internal ∼24 h rhythms to resonate with, and anticipate, external cycles of day and night. The mechanism underlying circadian entrainment to feeding time is critical for understanding why mistimed feeding, as occurs during shift work, disrupts circadian physiology, a state that is associated with increased incidence of chronic diseases such as type 2 (T2) diabetes. We show that feeding-regulated hormones insulin and insulin-like growth factor 1 (IGF-1) reset circadian clocks in vivo and in vitro by induction of PERIOD proteins, and mistimed insulin signaling disrupts circadian organization of mouse behavior and clock gene expression. Insulin and IGF-1 receptor signaling is sufficient to determine essential circadian parameters, principally via increased PERIOD protein synthesis. This requires coincident mechanistic target of rapamycin (mTOR) activation, increased phosphoinositide signaling, and microRNA downregulation. Besides its well-known homeostatic functions, we propose insulin and IGF-1 are primary signals of feeding time to cellular clocks throughout the body.


Subject(s)
Circadian Clocks/physiology , Feeding Behavior/physiology , Period Circadian Proteins/metabolism , Animals , Circadian Rhythm/physiology , Female , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Mammals/metabolism , Mice , Mice, Inbred C57BL , Receptor, IGF Type 1/metabolism , Signal Transduction
9.
Cell ; 177(6): 1583-1599.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150624

ABSTRACT

T and B cells are the two known lineages of adaptive immune cells. Here, we describe a previously unknown lymphocyte that is a dual expresser (DE) of TCR and BCR and key lineage markers of both B and T cells. In type 1 diabetes (T1D), DEs are predominated by one clonotype that encodes a potent CD4 T cell autoantigen in its antigen binding site. Molecular dynamics simulations revealed that this peptide has an optimal binding register for diabetogenic HLA-DQ8. In concordance, a synthetic version of the peptide forms stable DQ8 complexes and potently stimulates autoreactive CD4 T cells from T1D patients, but not healthy controls. Moreover, mAbs bearing this clonotype are autoreactive against CD4 T cells and inhibit insulin tetramer binding to CD4 T cells. Thus, compartmentalization of adaptive immune cells into T and B cells is not absolute, and violators of this paradigm are likely key drivers of autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Autoantigens/immunology , Child , Child, Preschool , Diabetes Mellitus, Type 1/metabolism , Epitopes/immunology , Female , HEK293 Cells , HLA-DQ Antigens/immunology , HLA-DQ Antigens/ultrastructure , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Middle Aged , Molecular Dynamics Simulation , Peptides , Protein Binding/immunology
10.
Cell ; 177(3): 722-736.e22, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30955890

ABSTRACT

Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.


Subject(s)
Gene Expression Regulation , Genome-Wide Association Study , Receptor, Insulin/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Regulation/drug effects , Host Cell Factor C1/antagonists & inhibitors , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , Protein Subunits/metabolism , RNA Interference , RNA Polymerase II/metabolism , RNA, Small Interfering/metabolism , Receptor, Insulin/chemistry , Signal Transduction/drug effects
11.
Immunity ; 57(2): 303-318.e6, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38309273

ABSTRACT

Production of amphiregulin (Areg) by regulatory T (Treg) cells promotes repair after acute tissue injury. Here, we examined the function of Treg cells in non-alcoholic steatohepatitis (NASH), a setting of chronic liver injury. Areg-producing Treg cells were enriched in the livers of mice and humans with NASH. Deletion of Areg in Treg cells, but not in myeloid cells, reduced NASH-induced liver fibrosis. Chronic liver damage induced transcriptional changes associated with Treg cell activation. Mechanistically, Treg cell-derived Areg activated pro-fibrotic transcriptional programs in hepatic stellate cells via epidermal growth factor receptor (EGFR) signaling. Deletion of Areg in Treg cells protected mice from NASH-dependent glucose intolerance, which also was dependent on EGFR signaling on hepatic stellate cells. Areg from Treg cells promoted hepatocyte gluconeogenesis through hepatocyte detection of hepatic stellate cell-derived interleukin-6. Our findings reveal a maladaptive role for Treg cell-mediated tissue repair functions in chronic liver disease and link liver damage to NASH-dependent glucose intolerance.


Subject(s)
Glucose Intolerance , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Amphiregulin/genetics , Amphiregulin/metabolism , ErbB Receptors/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Liver/metabolism , Liver Cirrhosis/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , T-Lymphocytes, Regulatory/metabolism
12.
Immunity ; 57(10): 2399-2415.e8, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39214091

ABSTRACT

T cell-mediated islet destruction is a hallmark of autoimmune diabetes. Here, we examined the dynamics and pathogenicity of CD4+ T cell responses to four different insulin-derived epitopes during diabetes initiation in non-obese diabetic (NOD) mice. Single-cell RNA sequencing of tetramer-sorted CD4+ T cells from the pancreas revealed that islet-antigen-specific T cells adopted a wide variety of fates and required XCR1+ dendritic cells for their activation. Hybrid-insulin C-chromogranin A (InsC-ChgA)-specific CD4+ T cells skewed toward a distinct T helper type 1 (Th1) effector phenotype, whereas the majority of insulin B chain and hybrid-insulin C-islet amyloid polypeptide-specific CD4+ T cells exhibited a regulatory phenotype and early or weak Th1 phenotype, respectively. InsC-ChgA-specific CD4+ T cells were uniquely pathogenic upon transfer, and an anti-InsC-ChgA:IAg7 antibody prevented spontaneous diabetes. Our findings highlight the heterogeneity of T cell responses to insulin-derived epitopes in diabetes and argue for the feasibility of antigen-specific therapies that blunts the response of pathogenic CD4+ T cells causing autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes , Chromogranin A , Diabetes Mellitus, Type 1 , Insulin , Mice, Inbred NOD , Animals , Diabetes Mellitus, Type 1/immunology , Chromogranin A/metabolism , Chromogranin A/immunology , Mice , Insulin/metabolism , Insulin/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Th1 Cells/immunology , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Peptides/immunology , Peptides/metabolism
13.
Immunity ; 57(6): 1289-1305.e9, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38772366

ABSTRACT

Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.


Subject(s)
Adipose Tissue , Homeostasis , Insulin Resistance , Lymphocytes , Mitochondria , Obesity , Programmed Cell Death 1 Receptor , Protein Serine-Threonine Kinases , Animals , Insulin Resistance/immunology , Programmed Cell Death 1 Receptor/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mitochondria/metabolism , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology , Obesity/immunology , Obesity/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Immunity, Innate , Male , Mitophagy/immunology , AMP-Activated Protein Kinase Kinases
14.
Cell ; 175(1): 146-158.e15, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30100182

ABSTRACT

Pathogen virulence exists on a continuum. The strategies that drive symptomatic or asymptomatic infections remain largely unknown. We took advantage of the concept of lethal dose 50 (LD50) to ask which component of individual non-genetic variation between hosts defines whether they survive or succumb to infection. Using the enteric pathogen Citrobacter, we found no difference in pathogen burdens between healthy and symptomatic populations. Iron metabolism-related genes were induced in asymptomatic hosts compared to symptomatic or naive mice. Dietary iron conferred complete protection without influencing pathogen burdens, even at 1000× the lethal dose of Citrobacter. Dietary iron induced insulin resistance, increasing glucose levels in the intestine that were necessary and sufficient to suppress pathogen virulence. A short course of dietary iron drove the selection of attenuated Citrobacter strains that can transmit and asymptomatically colonize naive hosts, demonstrating that environmental factors and cooperative metabolic strategies can drive conversion of pathogens toward commensalism.


Subject(s)
Host-Pathogen Interactions/physiology , Iron/metabolism , Virulence/physiology , Animals , Asymptomatic Infections , Citrobacter rodentium/metabolism , Citrobacter rodentium/pathogenicity , Colitis/drug therapy , Colitis/metabolism , Colon/microbiology , Dietary Supplements , Enterobacteriaceae Infections/drug therapy , Female , Insulin Resistance/physiology , Intestine, Small/microbiology , Iron/pharmacology , Lethal Dose 50 , Male , Mice , Mice, Inbred C3H , Mice, Inbred DBA
15.
Cell ; 172(4): 731-743.e12, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29425491

ABSTRACT

The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Energy Metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Transformed , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Oxygen Consumption/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , NF-kappaB-Inducing Kinase
16.
Annu Rev Cell Dev Biol ; 35: 501-521, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31590586

ABSTRACT

The dual leucine zipper-bearing kinase (DLK) and leucine zipper-bearing kinase (LZK) are evolutionarily conserved MAPKKKs of the mixed-lineage kinase family. Acting upstream of stress-responsive JNK and p38 MAP kinases, DLK and LZK have emerged as central players in neuronal responses to a variety of acute and traumatic injuries. Recent studies also implicate their function in astrocytes, microglia, and other nonneuronal cells, reflecting their expanding roles in the multicellular response to injury and in disease. Of particular note is the potential link of these kinases to neurodegenerative diseases and cancer. It is thus critical to understand the physiological contexts under which these kinases are activated, as well as the signal transduction mechanisms that mediate specific functional outcomes. In this review we first provide a historical overview of the biochemical and functional dissection of these kinases. We then discuss recent findings on regulating their activity to enhance cellular protection following injury and in disease, focusing on but not limited to the nervous system.


Subject(s)
Leucine Zippers/genetics , MAP Kinase Kinase Kinases/metabolism , Neurons/metabolism , Stress, Physiological/genetics , Animals , Axons/metabolism , Humans , MAP Kinase Kinase Kinases/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/virology , Neuroglia/metabolism , Neurons/virology , Regeneration/genetics , Regeneration/physiology , Stem Cells/metabolism , Stress, Physiological/physiology , Wounds and Injuries/genetics , Wounds and Injuries/metabolism
17.
Immunity ; 56(7): 1561-1577.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37402364

ABSTRACT

Hypodermis is the predominant site of Staphylococcus aureus infections that cause cellulitis. Given the importance of macrophages in tissue remodeling, we examined the hypodermal macrophages (HDMs) and their impact on host susceptibility to infection. Bulk and single-cell transcriptomics uncovered HDM subsets with CCR2-dichotomy. HDM homeostasis required the fibroblast-derived growth factor CSF1, ablation of which abrogated HDMs from the hypodermal adventitia. Loss of CCR2- HDMs resulted in accumulation of the extracellular matrix component, hyaluronic acid (HA). HDM-mediated HA clearance required sensing by the HA receptor, LYVE-1. Cell-autonomous IGF1 was required for accessibility of AP-1 transcription factor motifs that controlled LYVE-1 expression. Remarkably, loss of HDMs or IGF1 limited Staphylococcus aureus expansion via HA and conferred protection against cellulitis. Our findings reveal a function for macrophages in the regulation of HA with an impact on infection outcomes, which may be harnessed to limit the establishment of infection in the hypodermal niche.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/physiology , Cellulitis/metabolism , Macrophages/metabolism , Extracellular Matrix
18.
Cell ; 171(4): 824-835.e18, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29056338

ABSTRACT

Insulin resistance is a hallmark of diabetes and an unmet clinical need. Insulin inhibits hepatic glucose production and promotes lipogenesis by suppressing FOXO1-dependent activation of G6pase and inhibition of glucokinase, respectively. The tight coupling of these events poses a dual conundrum: mechanistically, as the FOXO1 corepressor of glucokinase is unknown, and clinically, as inhibition of glucose production is predicted to increase lipogenesis. Here, we report that SIN3A is the insulin-sensitive FOXO1 corepressor of glucokinase. Genetic ablation of SIN3A abolishes nutrient regulation of glucokinase without affecting other FOXO1 target genes and lowers glycemia without concurrent steatosis. To extend this work, we executed a small-molecule screen and discovered selective inhibitors of FOXO-dependent glucose production devoid of lipogenic activity in hepatocytes. In addition to identifying a novel mode of insulin action, these data raise the possibility of developing selective modulators of unliganded transcription factors to dial out adverse effects of insulin sensitizers.


Subject(s)
Forkhead Box Protein O1/antagonists & inhibitors , Glucose/metabolism , Hepatocytes/metabolism , Insulin Resistance , Acetylation , Animals , Cells, Cultured , Forkhead Box Protein O1/chemistry , Glucokinase/genetics , Glucokinase/metabolism , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , HEK293 Cells , Hepatocytes/enzymology , Histone Deacetylases/metabolism , Humans , Lipogenesis/drug effects , Mice , Mice, Knockout , Phosphorylation , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
19.
Cell ; 169(3): 470-482.e13, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431247

ABSTRACT

Aging is attended by a progressive decline in protein homeostasis (proteostasis), aggravating the risk for protein aggregation diseases. To understand the coordination between proteome imbalance and longevity, we addressed the mechanistic role of the quality-control ubiquitin ligase CHIP, which is a key regulator of proteostasis. We observed that CHIP deficiency leads to increased levels of the insulin receptor (INSR) and reduced lifespan of worms and flies. The membrane-bound INSR regulates the insulin and IGF1 signaling (IIS) pathway and thereby defines metabolism and aging. INSR is a direct target of CHIP, which triggers receptor monoubiquitylation and endocytic-lysosomal turnover to promote longevity. However, upon proteotoxic stress conditions and during aging, CHIP is recruited toward disposal of misfolded proteins, reducing its capacity to degrade the INSR. Our study indicates a competitive relationship between proteostasis and longevity regulation through CHIP-assisted proteolysis, providing a mechanistic concept for understanding the impact of proteome imbalance on aging.


Subject(s)
Aging , Antigens, CD/metabolism , Receptor, Insulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Caenorhabditis elegans , Drosophila melanogaster , Endocytosis , Humans , Longevity , Lysosomes/metabolism , Proteolysis , Proteome , Signal Transduction , Somatomedins , Ubiquitination
20.
Cell ; 171(2): 321-330.e14, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28965763

ABSTRACT

As organisms age, cells accumulate genetic and epigenetic errors that eventually lead to impaired organ function or catastrophic transformation such as cancer. Because aging reflects a stochastic process of increasing disorder, cells in an organ will be individually affected in different ways, thus rendering bulk analyses of postmitotic adult cells difficult to interpret. Here, we directly measure the effects of aging in human tissue by performing single-cell transcriptome analysis of 2,544 human pancreas cells from eight donors spanning six decades of life. We find that islet endocrine cells from older donors display increased levels of transcriptional noise and potential fate drift. By determining the mutational history of individual cells, we uncover a novel mutational signature in healthy aging endocrine cells. Our results demonstrate the feasibility of using single-cell RNA sequencing (RNA-seq) data from primary cells to derive insights into genetic and transcriptional processes that operate on aging human tissue.


Subject(s)
Aging/pathology , Cellular Senescence , Mutation , Pancreas/pathology , Single-Cell Analysis , Adult , Child , Child, Preschool , Humans , Infant , Middle Aged , Pancreas/cytology , Pancreas/physiology , Polymorphism, Single Nucleotide , Sequence Analysis, RNA , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL