Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138.762
Filter
Add more filters

Publication year range
1.
Cell ; 187(1): 204-215.e14, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38070508

ABSTRACT

Mounting evidence suggests metabolism instructs stem cell fate decisions. However, how fetal metabolism changes during development and how altered maternal metabolism shapes fetal metabolism remain unexplored. We present a descriptive atlas of in vivo fetal murine metabolism during mid-to-late gestation in normal and diabetic pregnancy. Using 13C-glucose and liquid chromatography-mass spectrometry (LC-MS), we profiled the metabolism of fetal brains, hearts, livers, and placentas harvested from pregnant dams between embryonic days (E)10.5 and 18.5. Our analysis revealed metabolic features specific to a hyperglycemic environment and signatures that may denote developmental transitions during euglycemic development. We observed sorbitol accumulation in fetal tissues and altered neurotransmitter levels in fetal brains isolated from hyperglycemic dams. Tracing 13C-glucose revealed disparate fetal nutrient sourcing depending on maternal glycemic states. Regardless of glycemic state, histidine-derived metabolites accumulated in late-stage fetal tissues. Our rich dataset presents a comprehensive overview of in vivo fetal tissue metabolism and alterations due to maternal hyperglycemia.


Subject(s)
Diabetes Mellitus , Diabetes, Gestational , Fetus , Animals , Female , Mice , Pregnancy , Diabetes Mellitus/metabolism , Fetus/metabolism , Glucose/metabolism , Placenta/metabolism , Diabetes, Gestational/metabolism
2.
Cell ; 168(1-2): 73-85.e11, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916274

ABSTRACT

The recent discovery that genetically modified α cells can regenerate and convert into ß-like cells in vivo holds great promise for diabetes research. However, to eventually translate these findings to human, it is crucial to discover compounds with similar activities. Herein, we report the identification of GABA as an inducer of α-to-ß-like cell conversion in vivo. This conversion induces α cell replacement mechanisms through the mobilization of duct-lining precursor cells that adopt an α cell identity prior to being converted into ß-like cells, solely upon sustained GABA exposure. Importantly, these neo-generated ß-like cells are functional and can repeatedly reverse chemically induced diabetes in vivo. Similarly, the treatment of transplanted human islets with GABA results in a loss of α cells and a concomitant increase in ß-like cell counts, suggestive of α-to-ß-like cell conversion processes also in humans. This newly discovered GABA-induced α cell-mediated ß-like cell neogenesis could therefore represent an unprecedented hope toward improved therapies for diabetes.


Subject(s)
Diabetes Mellitus/drug therapy , Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , gamma-Aminobutyric Acid/administration & dosage , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucagon-Secreting Cells/drug effects , Humans , Islets of Langerhans/cytology , Male , Mice , Nerve Tissue Proteins , Rats , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology
3.
Cell ; 168(6): 960-976, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283069

ABSTRACT

The mechanistic target of rapamycin (mTOR) coordinates eukaryotic cell growth and metabolism with environmental inputs, including nutrients and growth factors. Extensive research over the past two decades has established a central role for mTOR in regulating many fundamental cell processes, from protein synthesis to autophagy, and deregulated mTOR signaling is implicated in the progression of cancer and diabetes, as well as the aging process. Here, we review recent advances in our understanding of mTOR function, regulation, and importance in mammalian physiology. We also highlight how the mTOR signaling network contributes to human disease and discuss the current and future prospects for therapeutically targeting mTOR in the clinic.


Subject(s)
Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Aging/metabolism , Animals , Diabetes Mellitus/metabolism , Glucose/metabolism , Humans , Muscles/metabolism , Neoplasms/metabolism
4.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916275

ABSTRACT

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Subject(s)
Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
5.
Cell ; 160(5): 816-827, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723161

ABSTRACT

While modernization has dramatically increased lifespan, it has also witnessed the increasing prevalence of diseases such as obesity, hypertension, and type 2 diabetes. Such chronic, acquired diseases result when normal physiologic control goes awry and may thus be viewed as failures of homeostasis. However, while nearly every process in human physiology relies on homeostatic mechanisms for stability, only some have demonstrated vulnerability to dysregulation. Additionally, chronic inflammation is a common accomplice of the diseases of homeostasis, yet the basis for this connection is not fully understood. Here we review the design of homeostatic systems and discuss universal features of control circuits that operate at the cellular, tissue, and organismal levels. We suggest a framework for classification of homeostatic signals that is based on different classes of homeostatic variables they report on. Finally, we discuss how adaptability of homeostatic systems with adjustable set points creates vulnerability to dysregulation and disease. This framework highlights the fundamental parallels between homeostatic and inflammatory control mechanisms and provides a new perspective on the physiological origin of inflammation.


Subject(s)
Disease Susceptibility , Homeostasis , Inflammation/physiopathology , Diabetes Mellitus/metabolism , Humans , Models, Biological
6.
Cell ; 163(6): 1457-67, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26627735

ABSTRACT

A variety of signals finely tune insulin secretion by pancreatic ß cells to prevent both hyper-and hypoglycemic states. Here, we show that post-translational regulation of the transcription factors ETV1, ETV4, and ETV5 by the ubiquitin ligase COP1 (also called RFWD2) in ß cells is critical for insulin secretion. Mice lacking COP1 in ß cells developed diabetes due to insulin granule docking defects that were fully rescued by genetic deletion of Etv1, Etv4, and Etv5. Genes regulated by ETV1, ETV4, or ETV5 in the absence of mouse COP1 were enriched in human diabetes-associated genes, suggesting that they also influence human ß-cell pathophysiology. In normal ß cells, ETV4 was stabilized upon membrane depolarization and limited insulin secretion under hyperglycemic conditions. Collectively, our data reveal that ETVs negatively regulate insulin secretion for the maintenance of normoglycemia.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , DNA-Binding Proteins/metabolism , Diabetes Mellitus/metabolism , Exocytosis , Gene Deletion , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Insulin Secretion , Mice , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
7.
Nature ; 625(7994): 312-320, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200293

ABSTRACT

The Holocene (beginning around 12,000 years ago) encompassed some of the most significant changes in human evolution, with far-reaching consequences for the dietary, physical and mental health of present-day populations. Using a dataset of more than 1,600 imputed ancient genomes1, we modelled the selection landscape during the transition from hunting and gathering, to farming and pastoralism across West Eurasia. We identify key selection signals related to metabolism, including that selection at the FADS cluster began earlier than previously reported and that selection near the LCT locus predates the emergence of the lactase persistence allele by thousands of years. We also find strong selection in the HLA region, possibly due to increased exposure to pathogens during the Bronze Age. Using ancient individuals to infer local ancestry tracts in over 400,000 samples from the UK Biobank, we identify widespread differences in the distribution of Mesolithic, Neolithic and Bronze Age ancestries across Eurasia. By calculating ancestry-specific polygenic risk scores, we show that height differences between Northern and Southern Europe are associated with differential Steppe ancestry, rather than selection, and that risk alleles for mood-related phenotypes are enriched for Neolithic farmer ancestry, whereas risk alleles for diabetes and Alzheimer's disease are enriched for Western hunter-gatherer ancestry. Our results indicate that ancient selection and migration were large contributors to the distribution of phenotypic diversity in present-day Europeans.


Subject(s)
Asian , European People , Genome, Human , Selection, Genetic , Humans , Affect , Agriculture/history , Alleles , Alzheimer Disease/genetics , Asia/ethnology , Asian/genetics , Diabetes Mellitus/genetics , Europe/ethnology , European People/genetics , Farmers/history , Genetic Loci/genetics , Genetic Predisposition to Disease , Genome, Human/genetics , History, Ancient , Human Migration , Hunting/history , Multigene Family/genetics , Phenotype , UK Biobank , Multifactorial Inheritance/genetics
8.
Immunity ; 53(4): 864-877.e5, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32791036

ABSTRACT

The SARS-CoV-2 pandemic has resulted in millions of infections, yet the role of host immune responses in early COVID-19 pathogenesis remains unclear. By investigating 17 acute and 24 convalescent patients, we found that acute SARS-CoV-2 infection resulted in broad immune cell reduction including T, natural killer, monocyte, and dendritic cells (DCs). DCs were significantly reduced with functional impairment, and ratios of conventional DCs to plasmacytoid DCs were increased among acute severe patients. Besides lymphocytopenia, although neutralizing antibodies were rapidly and abundantly generated in patients, there were delayed receptor binding domain (RBD)- and nucleocapsid protein (NP)-specific T cell responses during the first 3 weeks after symptoms onset. Moreover, acute RBD- and NP-specific T cell responses included relatively more CD4 T cells than CD8 T cells. Our findings provided evidence that impaired DCs, together with timely inverted strong antibody but weak CD8 T cell responses, could contribute to acute COVID-19 pathogenesis and have implications for vaccine development.


Subject(s)
Betacoronavirus/pathogenicity , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Dendritic Cells/immunology , Diabetes Mellitus/immunology , Hypertension/immunology , Pneumonia, Viral/immunology , Adult , Aged , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Betacoronavirus/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , COVID-19 , Convalescence , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Dendritic Cells/pathology , Dendritic Cells/virology , Diabetes Complications , Diabetes Mellitus/diagnosis , Diabetes Mellitus/virology , Disease Progression , Female , Humans , Hypertension/complications , Hypertension/diagnosis , Hypertension/virology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lymphocyte Activation , Lymphocyte Count , Male , Middle Aged , Monocytes/immunology , Monocytes/pathology , Monocytes/virology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index
9.
Cell ; 159(4): 911-24, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417165

ABSTRACT

The development of whole-body imaging at single-cell resolution enables system-level approaches to studying cellular circuits in organisms. Previous clearing methods focused on homogenizing mismatched refractive indices of individual tissues, enabling reductions in opacity but falling short of achieving transparency. Here, we show that an aminoalcohol decolorizes blood by efficiently eluting the heme chromophore from hemoglobin. Direct transcardial perfusion of an aminoalcohol-containing cocktail that we previously termed CUBIC coupled with a 10 day to 2 week clearing protocol decolorized and rendered nearly transparent almost all organs of adult mice as well as the entire body of infant and adult mice. This CUBIC-perfusion protocol enables rapid whole-body and whole-organ imaging at single-cell resolution by using light-sheet fluorescent microscopy. The CUBIC protocol is also applicable to 3D pathology, anatomy, and immunohistochemistry of various organs. These results suggest that whole-body imaging of colorless tissues at high resolution will contribute to organism-level systems biology.


Subject(s)
Amino Alcohols/analysis , Single-Cell Analysis/methods , Whole Body Imaging/methods , Animals , Diabetes Mellitus/pathology , Imaging, Three-Dimensional/methods , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL
10.
Nature ; 624(7992): 645-652, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38093014

ABSTRACT

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Subject(s)
Dendritic Cells , Diabetes Complications , Diabetes Mellitus , Disease Susceptibility , Hyperglycemia , Lung , Virus Diseases , Animals , Mice , Acetyl Coenzyme A/metabolism , Acetylation , Chromatin/genetics , Chromatin/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diabetes Complications/immunology , Diabetes Complications/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glucose/metabolism , Histones/metabolism , Hyperglycemia/complications , Hyperglycemia/immunology , Hyperglycemia/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , T-Lymphocytes/immunology , Virus Diseases/complications , Virus Diseases/immunology , Virus Diseases/mortality , Viruses/immunology , Disease Models, Animal , Humans
11.
Mol Cell ; 81(3): 629-637.e5, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33400924

ABSTRACT

As a master regulator of metabolism, AMP-activated protein kinase (AMPK) is activated upon energy and glucose shortage but suppressed upon overnutrition. Exaggerated negative regulation of AMPK signaling by nutrient overload plays a crucial role in metabolic diseases. However, the mechanism underlying the negative regulation is poorly understood. Here, we demonstrate that high glucose represses AMPK signaling via MG53 (also called TRIM72) E3-ubiquitin-ligase-mediated AMPKα degradation and deactivation. Specifically, high-glucose-stimulated reactive oxygen species (ROS) signals AKT to phosphorylate AMPKα at S485/491, which facilitates the recruitment of MG53 and the subsequent ubiquitination and degradation of AMPKα. In addition, high glucose deactivates AMPK by ROS-dependent suppression of phosphorylation of AMPKα at T172. These findings not only delineate the mechanism underlying the impairment of AMPK signaling in overnutrition-related diseases but also highlight the significance of keeping the yin-yang balance of AMPK signaling in the maintenance of metabolic homeostasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/enzymology , Glucose/pharmacology , Membrane Proteins/metabolism , Muscle, Skeletal/drug effects , Obesity/enzymology , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/genetics , Animals , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/genetics , Disease Models, Animal , HEK293 Cells , Humans , Macaca mulatta , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Muscle, Skeletal/enzymology , Obesity/blood , Obesity/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Reactive Oxygen Species/metabolism , Signal Transduction , Ubiquitination
12.
Trends Biochem Sci ; 49(2): 99-100, 2024 02.
Article in English | MEDLINE | ID: mdl-37770288

ABSTRACT

Wang et al. identified dipeptidyl peptidase 4 (DPP4) as a gut microbe-derived enzyme that impacts on host glucose metabolism. They further introduced a novel therapeutic, daurisoline-d4 (Dau-d4), a selective microbial DPP4 (mDPP4) inhibitor that shows promise in improving glucose tolerance, highlighting the potential of therapies that target both host enzymes and gut microbial enzymes.


Subject(s)
Diabetes Mellitus , Dipeptidyl-Peptidase IV Inhibitors , Gastrointestinal Microbiome , Humans , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use
13.
Nat Immunol ; 17(3): 297-303, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26752376

ABSTRACT

Inflammation induced during infection can both promote and suppress immunity. This contradiction suggests that inflammatory cytokines affect the immune system in a context-dependent manner. Here we show that nonspecific bystander inflammation conditions naive CD4(+) T cells for enhanced peripheral Foxp3 induction and reduced effector differentiation. This results in inhibition of immune responses in vivo via a Foxp3-dependent effect on antigen-specific naive CD4(+) T cell precursors. Such conditioning may have evolved to allow immunity to infection while limiting subsequent autoimmunity caused by release of self-antigens in the wake of infection. Furthermore, this phenomenon suggests a mechanistic explanation for the idea that early tuning of the immune system by infection affects the long-term quality of immune regulation.


Subject(s)
Asthma/immunology , Autoimmunity/immunology , Bystander Effect/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Diabetes Mellitus/immunology , Forkhead Transcription Factors/immunology , Inflammation , Self Tolerance/immunology , Animals , Autoantigens , Bystander Effect/drug effects , CD4-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Cytokines/drug effects , Cytokines/pharmacology , DNA Methylation , Disease Models, Animal , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Interferon Inducers/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peripheral Tolerance/immunology , Poly I-C/pharmacology , Promoter Regions, Genetic , Thymic Stromal Lymphopoietin
14.
Nature ; 606(7915): 776-784, 2022 06.
Article in English | MEDLINE | ID: mdl-35614212

ABSTRACT

Chronic non-healing wounds are a major complication of diabetes, which affects 1 in 10 people worldwide. Dying cells in the wound perpetuate the inflammation and contribute to dysregulated tissue repair1-3. Here we reveal that the membrane transporter SLC7A11 acts as a molecular brake on efferocytosis, the process by which dying cells are removed, and that inhibiting SLC7A11 function can accelerate wound healing. Transcriptomics of efferocytic dendritic cells in mouse identified upregulation of several SLC7 gene family members. In further analyses, pharmacological inhibition of SLC7A11, or deletion or knockdown of Slc7a11 using small interfering RNA enhanced efferocytosis in dendritic cells. Slc7a11 was highly expressed in dendritic cells in skin, and single-cell RNA sequencing of inflamed skin showed that Slc7a11 was upregulated in innate immune cells. In a mouse model of excisional skin wounding, inhibition or loss of SLC7A11 expression accelerated healing dynamics and reduced the apoptotic cell load in the wound. Mechanistic studies revealed a link between SLC7A11, glucose homeostasis and diabetes. SLC7A11-deficient dendritic cells were dependent on aerobic glycolysis using glucose derived from glycogen stores for increased efferocytosis; also, transcriptomics of efferocytic SLC7A11-deficient dendritic cells identified increased expression of genes linked to gluconeogenesis and diabetes. Further, Slc7a11 expression was higher in the wounds of diabetes-prone db/db mice, and targeting SLC7A11 accelerated their wound healing. The faster healing was also linked to the release of the TGFß family member GDF15 from efferocytic dendritic cells. In sum, SLC7A11 is a negative regulator of efferocytosis, and removing this brake improves wound healing, with important implications for wound management in diabetes.


Subject(s)
Amino Acid Transport System y+ , Dendritic Cells , Diabetes Mellitus , Phagocytosis , Wound Healing , Amino Acid Transport System y+/antagonists & inhibitors , Animals , Dendritic Cells/cytology , Dendritic Cells/immunology , Diabetes Mellitus/immunology , Gluconeogenesis , Glucose , Glycolysis , Growth Differentiation Factor 15 , Mice
15.
Nature ; 603(7899): 159-165, 2022 03.
Article in English | MEDLINE | ID: mdl-35197629

ABSTRACT

Metformin, the most prescribed antidiabetic medicine, has shown other benefits such as anti-ageing and anticancer effects1-4. For clinical doses of metformin, AMP-activated protein kinase (AMPK) has a major role in its mechanism of action4,5; however, the direct molecular target of metformin remains unknown. Here we show that clinically relevant concentrations of metformin inhibit the lysosomal proton pump v-ATPase, which is a central node for AMPK activation following glucose starvation6. We synthesize a photoactive metformin probe and identify PEN2, a subunit of γ-secretase7, as a binding partner of metformin with a dissociation constant at micromolar levels. Metformin-bound PEN2 forms a complex with ATP6AP1, a subunit of the v-ATPase8, which leads to the inhibition of v-ATPase and the activation of AMPK without effects on cellular AMP levels. Knockout of PEN2 or re-introduction of a PEN2 mutant that does not bind ATP6AP1 blunts AMPK activation. In vivo, liver-specific knockout of Pen2 abolishes metformin-mediated reduction of hepatic fat content, whereas intestine-specific knockout of Pen2 impairs its glucose-lowering effects. Furthermore, knockdown of pen-2 in Caenorhabditis elegans abrogates metformin-induced extension of lifespan. Together, these findings reveal that metformin binds PEN2 and initiates a signalling route that intersects, through ATP6AP1, the lysosomal glucose-sensing pathway for AMPK activation. This ensures that metformin exerts its therapeutic benefits in patients without substantial adverse effects.


Subject(s)
Hypoglycemic Agents , Metformin , Vacuolar Proton-Translocating ATPases , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphatases/metabolism , Amyloid Precursor Protein Secretases , Animals , Caenorhabditis elegans/metabolism , Diabetes Mellitus/drug therapy , Glucose/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Lysosomes/metabolism , Membrane Proteins , Metformin/agonists , Metformin/metabolism , Metformin/pharmacology , Vacuolar Proton-Translocating ATPases/metabolism
16.
Annu Rev Biochem ; 81: 507-32, 2012.
Article in English | MEDLINE | ID: mdl-22482906

ABSTRACT

To enhance glucose uptake into muscle and fat cells, insulin stimulates the translocation of GLUT4 glucose transporters from intracellular membranes to the cell surface. This response requires the intersection of insulin signaling and vesicle trafficking pathways, and it is compromised in the setting of overnutrition to cause insulin resistance. Insulin signals through AS160/Tbc1D4 and Tbc1D1 to modulate Rab GTPases and through the Rho GTPase TC10α to act on other targets. In unstimulated cells, GLUT4 is incorporated into specialized storage vesicles containing IRAP, LRP1, sortilin, and VAMP2, which are sequestered by TUG, Ubc9, and other proteins. Insulin mobilizes these vesicles directly to the plasma membrane, and it modulates the trafficking itinerary so that cargo recycles from endosomes during ongoing insulin exposure. Knowledge of how signaling and trafficking pathways are coordinated will be essential to understanding the pathogenesis of diabetes and the metabolic syndrome and may also inform a wide range of other physiologies.


Subject(s)
Diabetes Mellitus/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Insulin/metabolism , Signal Transduction , Animals , Glucose/metabolism , Humans
17.
Genes Dev ; 34(23-24): 1559-1561, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33262142

ABSTRACT

In vivo regeneration of ß cells provides hope for self-renewal of functional insulin-secreting cells following ß-cell failure, a historically fatal condition now sustainable only by administration of exogenous insulin. Despite advances in the treatment of diabetes mellitus, the path toward endogenous renewal of ß-cell populations has remained elusive. Intensive efforts have focused on elucidating pancreatic transcriptional programs that can drive the division and (trans-)differentiation of non-ß cells to produce insulin. A surprise has been the identification of an essential role of the molecular circadian clock in the regulation of competent insulin-producing ß cells. In this issue of Genes & Development, work by Petrenko and colleagues (pp. 1650-1665) now shows a requirement for the intrinsic clock in the regenerative capacity of insulin-producing cells following genetic ablation of ß cells. These studies raise the possibility that enhancing core clock activity may provide an adjuvant in cell replacement therapies.


Subject(s)
Circadian Clocks , Diabetes Mellitus , Insulin-Secreting Cells , Humans , Insulin , Pancreas
18.
Physiol Rev ; 100(4): 1599-1619, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32666886

ABSTRACT

Sentrin/small ubiquitin-like modifier (SUMO) is protein modification pathway that regulates multiple biological processes, including cell division, DNA replication/repair, signal transduction, and cellular metabolism. In this review, we will focus on recent advances in the mechanisms of disease pathogenesis, such as cancer, diabetes, seizure, and heart failure, which have been linked to the SUMO pathway. SUMO is conjugated to lysine residues in target proteins through an isopeptide linkage catalyzed by SUMO-specific activating (E1), conjugating (E2), and ligating (E3) enzymes. In steady state, the quantity of SUMO-modified substrates is usually a small fraction of unmodified substrates due to the deconjugation activity of the family Sentrin/SUMO-specific proteases (SENPs). In contrast to the complexity of the ubiquitination/deubiquitination machinery, the biochemistry of SUMOylation and de-SUMOylation is relatively modest. Specificity of the SUMO pathway is achieved through redox regulation, acetylation, phosphorylation, or other posttranslational protein modification of the SUMOylation and de-SUMOylation enzymes. There are three major SUMOs. SUMO-1 usually modifies a substrate as a monomer; however, SUMO-2/3 can form poly-SUMO chains. The monomeric SUMO-1 or poly-SUMO chains can interact with other proteins through SUMO-interactive motif (SIM). Thus SUMO modification provides a platform to enhance protein-protein interaction. The consequence of SUMOylation includes changes in cellular localization, protein activity, or protein stability. Furthermore, SUMO may join force with ubiquitin to degrade proteins through SUMO-targeted ubiquitin ligases (STUbL). After 20 yr of research, SUMO has been shown to play critical roles in most, if not all, biological pathways. Thus the SUMO enzymes could be targets for drug development to treat human diseases.


Subject(s)
Small Ubiquitin-Related Modifier Proteins/metabolism , Adaptive Immunity/physiology , Animals , Cardiovascular Diseases/metabolism , Diabetes Mellitus/metabolism , Humans , Neoplasms/metabolism , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/genetics
19.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38265192

ABSTRACT

The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.


Subject(s)
Diabetes Mellitus , Islets of Langerhans , Humans , Cell Differentiation , Organogenesis , Pancreas
20.
Immunity ; 49(1): 164-177.e6, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958802

ABSTRACT

Pro-inflammatory cytokines of a T helper-1-signature are known to promote insulin resistance (IR) in obesity, but the physiological role of this mechanism is unclear. It is also unknown whether and how viral infection induces loss of glycemic control in subjects at risk for developing diabetes mellitus type 2 (DM2). We have found in mice and humans that viral infection caused short-term systemic IR. Virally-induced interferon-γ (IFN-γ) directly targeted skeletal muscle to downregulate the insulin receptor but did not cause loss of glycemic control because of a compensatory increase of insulin production. Hyperinsulinemia enhanced antiviral immunity through direct stimulation of CD8+ effector T cell function. In pre-diabetic mice with hepatic IR caused by diet-induced obesity, infection resulted in loss of glycemic control. Thus, upon pathogen encounter, the immune system transiently reduces insulin sensitivity of skeletal muscle to induce hyperinsulinemia and promote antiviral immunity, which derails to glucose intolerance in pre-diabetic obese subjects. VIDEO ABSTRACT.


Subject(s)
Diabetes Mellitus/immunology , Insulin Resistance , Insulin/immunology , Interferon-gamma/metabolism , Muscle, Skeletal/metabolism , Obesity/immunology , Virus Diseases/complications , Animals , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/virology , Disease Models, Animal , Down-Regulation/genetics , Humans , Hyperinsulinism , Insulin/blood , Male , Mice , Obesity/complications , Obesity/metabolism , Obesity/virology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL