Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Metab ; 35(12): 2200-2215.e9, 2023 12 05.
Article in English | MEDLINE | ID: mdl-37949065

ABSTRACT

During the progression of type 1 diabetes (T1D), ß cells are exposed to significant stress and, therefore, require adaptive responses to survive. The adaptive mechanisms that can preserve ß cell function and survival in the face of autoimmunity remain unclear. Here, we show that the deletion of the unfolded protein response (UPR) genes Atf6α or Ire1α in ß cells of non-obese diabetic (NOD) mice prior to insulitis generates a p21-driven early senescence phenotype and alters the ß cell secretome that significantly enhances the leukemia inhibitory factor-mediated recruitment of M2 macrophages to islets. Consequently, M2 macrophages promote anti-inflammatory responses and immune surveillance that cause the resolution of islet inflammation, the removal of terminally senesced ß cells, the reduction of ß cell apoptosis, and protection against T1D. We further demonstrate that the p21-mediated early senescence signature is conserved in the residual ß cells of T1D patients. Our findings reveal a previously unrecognized link between ß cell UPR and senescence that, if leveraged, may represent a novel preventive strategy for T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Animals , Humans , Diabetes Mellitus, Type 1/metabolism , Endoribonucleases/metabolism , Mice, Inbred NOD , Protein Serine-Threonine Kinases/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism
2.
Mol Metab ; 54: 101365, 2021 12.
Article in English | MEDLINE | ID: mdl-34728341

ABSTRACT

BACKGROUND: Pancreatic ß-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, ß-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to ß-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of ß-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of ß-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW: In this article, we introduce ER stress and the UPR, review ß-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on ß-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS: Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that ß-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of ß-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in ß-cells and uncovering the crosstalk between stressed ß-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Animals , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Stress , Humans
3.
Sci Signal ; 14(683)2021 05 18.
Article in English | MEDLINE | ID: mdl-34006608

ABSTRACT

Developing hippocampal neurons undergo rapid synaptogenesis in response to neurotrophic signals to form and refine circuit connections. The adipokine leptin is a satiety factor with neurotrophic actions, which potentiates both glutamatergic and GABAergic synaptogenesis in the hippocampus during neonatal development. Brief exposure to leptin enhances GABAA receptor-dependent synaptic currents in hippocampal neurons. Here, using molecular and electrophysiological techniques, we found that leptin increased the surface localization of GABAA receptors and the number of functional GABAergic synapses in hippocampal cultures from male and female rat pups. Leptin increased the interaction between GABAA receptors and the Rho guanine exchange factor ß-PIX (a scaffolding protein at GABAergic postsynaptic sites) in a manner dependent on the kinase CaMKK. We also found that the leptin receptor and ß-PIX formed a complex, the amount of which transiently increased upon leptin receptor activation. Furthermore, Tyr985 in the leptin receptor and the SH3 domain of ß-PIX are crucial for this interaction, which was required for the developmental increase in GABAergic synaptogenesis. Our results suggest a mechanism by which leptin promotes GABAergic synaptogenesis in hippocampal neurons and reveal further complexity in leptin receptor signaling and its interactome.


Subject(s)
Leptin , Neurons , Rho Guanine Nucleotide Exchange Factors , Animals , Female , Hippocampus/cytology , Leptin/metabolism , Male , Neurons/metabolism , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Synapses/metabolism
4.
Mol Cell Neurosci ; 106: 103500, 2020 07.
Article in English | MEDLINE | ID: mdl-32438059

ABSTRACT

Normal development of neuronal connections in the hippocampus requires neurotrophic signals, including the cytokine leptin. During neonatal development, leptin induces formation and maturation of dendritic spines, the main sites of glutamatergic synapses in the hippocampal neurons. However, the molecular mechanisms for leptin-induced synaptogenesis are not entirely understood. In this study, we reveal two novel targets of leptin in developing hippocampal neurons and address their role in synaptogenesis. First target is Kruppel-Like Factor 4 (KLF4), which we identified using a genome-wide target analysis strategy. We show that leptin upregulates KLF4 in hippocampal neurons and that leptin signaling is important for KLF4 expression in vivo. Furthermore, KLF4 is required for leptin-induced synaptogenesis, as shKLF4 blocks and upregulation of KLF4 phenocopies it. We go on to show that KLF4 requires its signal transducer and activator of transcription 3 (STAT3) binding site and thus potentially blocks STAT3 activity to induce synaptogenesis. Second, we show that leptin increases the expression of suppressor of cytokine signaling 3 (SOCS3), another well-known inhibitor of STAT3, in developing hippocampal neurons. SOCS3 is also required for leptin-induced synaptogenesis and sufficient to stimulate it alone. Finally, we show that constitutively active STAT3 blocks the effects of leptin on spine formation, while the targeted knockdown of STAT3 is sufficient to induce it. Overall, our data demonstrate that leptin increases the expression of both KLF4 and SOCS3, inhibiting the activity of STAT3 in the hippocampal neurons and resulting in the enhancement of glutamatergic synaptogenesis during neonatal development.


Subject(s)
Hippocampus/drug effects , Leptin/pharmacology , Neurons/drug effects , Signal Transduction/drug effects , Synapses/drug effects , Animals , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Female , Hippocampus/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Neurogenesis/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Synapses/metabolism , Transcriptome
5.
Endocrinology ; 161(2)2020 02 01.
Article in English | MEDLINE | ID: mdl-31840160

ABSTRACT

Activation of the leptin receptor, LepRb, by the adipocytokine/neurotrophic factor leptin in the central nervous system has procognitive and antidepressive effects. Leptin has been shown to increase glutamatergic synaptogenesis in multiple brain regions. In contrast, mice that have a mutation in the LepRb gene show abnormal synapse development in the hippocampus as well as deficits in cognition and increased depressive-like symptoms. Leptin increases glutamatergic synaptogenesis, in part, through enhancement of N-methyl-D-aspartic acid (NMDA) receptor function; yet the underlying signaling pathway is not known. In this study, we examine how leptin regulates surface expression of NR2B-containing NMDA receptors in hippocampal neurons. Leptin stimulation increases NR2BY1472 phosphorylation, which is inhibited by the Src family kinase inhibitor, PP1. Moreover, we show that Fyn, a member of the Src family kinases, is required for leptin-stimulated NR2BY1472 phosphorylation. Furthermore, inhibiting Y1472 phosphorylation with either a dominant negative Fyn mutant or an NR2B mutant that lacks the phosphorylation site (NR2BY1472F) blocks leptin-stimulated synaptogenesis. Additionally, we show that LepRb forms a complex with NR2B and Fyn. Taken together, these findings expand our knowledge of the LepRb interactome and the mechanisms by which leptin stimulates glutamatergic synaptogenesis in the developing hippocampus. Comprehending these mechanisms is key for understanding dendritic spine development and synaptogenesis, alterations of which are associated with many neurological disorders.


Subject(s)
Hippocampus/physiology , Leptin/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Leptin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Animals , HEK293 Cells , Humans , Phosphorylation , Primary Cell Culture , Rats
6.
Endocrinology ; 160(8): 1982-1998, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31199479

ABSTRACT

Leptin has neurotrophic actions in the hippocampus to increase synapse formation and stimulate neuronal plasticity. Leptin also enhances cognition and has antidepressive and anxiolytic-like effects, two hippocampal-dependent behaviors. In contrast, mice lacking leptin or the long form of the leptin receptor (LepRb) have lower cortical volume and decreased memory and exhibit depressive-like behaviors. A number of the signaling pathways regulated by LepRb are known, but how membrane LepRb levels are regulated in the central nervous system is not well understood. Here, we show that the lysosomal inhibitor chloroquine increases LepRb expression in hippocampal cultures, suggesting that LepRb is degraded in the lysosome. Furthermore, we show that leptin increases surface expression of its own receptor by decreasing the level of ubiquitinated LepRbs. This decrease is mediated by the deubiquitinase ubiquitin-specific protease 8 (USP8), which we show is in complex with LepRb. Acute leptin stimulation increases USP8 activity. Moreover, leptin stimulates USP8 gene expression through cAMP response element-binding protein (CREB)-dependent transcription, an effect blocked by expression of a dominant-negative CREB or with short hairpin RNA knockdown of CREB. Increased expression of USP8 causes increased surface localization of LepRb, which in turn enhances leptin-mediated activation of the MAPK kinase/extracellular signal-regulated kinase pathway and CREB activation. Lastly, increased USP8 expression increases glutamatergic synapse formation in hippocampal cultures, an effect dependent on expression of LepRbs. Leptin-stimulated synapse formation also requires USP8. In conclusion, we show that USP8 deubiquitinates LepRb, thus inhibiting lysosomal degradation and enhancing surface localization of LepRb, which are essential for leptin-stimulated synaptogenesis in the hippocampus.


Subject(s)
Endopeptidases/physiology , Endosomal Sorting Complexes Required for Transport/physiology , Leptin/pharmacology , Receptors, Leptin/metabolism , Synapses/physiology , Ubiquitin Thiolesterase/physiology , Ubiquitination , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/physiology , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Ubiquitin Thiolesterase/genetics
7.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28303257

ABSTRACT

Calcium signaling controls many key processes in neurons, including gene expression, axon guidance, and synaptic plasticity. In contrast to calcium influx through voltage- or neurotransmitter-gated channels, regulatory pathways that control store-operated calcium entry (SOCE) in neurons are poorly understood. Here, we report a transcriptional control of Stim1 (stromal interaction molecule 1) gene, which is a major sensor of endoplasmic reticulum (ER) calcium levels and a regulator of SOCE. By using a genome-wide chromatin immunoprecipitation and sequencing approach in mice, we find that NEUROD2, a neurogenic transcription factor, binds to an intronic element within the Stim1 gene. We show that NEUROD2 limits Stim1 expression in cortical neurons and consequently fine-tunes the SOCE response upon depletion of ER calcium. Our findings reveal a novel mechanism that regulates neuronal calcium homeostasis during cortical development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Stromal Interaction Molecule 1/metabolism , Animals , Animals, Newborn , Cell Cycle Proteins , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Conserved Sequence , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Introns , Mice, Inbred BALB C , Nuclear Proteins , Protein Binding , Transcription Factors
8.
BMC Genomics ; 16: 681, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-26341353

ABSTRACT

BACKGROUND: Cellular differentiation programs are controlled, to a large extent, by the combinatorial functioning of specific transcription factors. Cortical projection neurons constitute the major excitatory neuron population within the cortex and mediate long distance communication between the cortex and other brain regions. Our understanding of effector transcription factors and their downstream transcriptional programs that direct the differentiation process of cortical projection neurons is far from complete. RESULTS: In this study, we carried out a ChIP-Seq (chromatin-immunoprecipitation and sequencing) analysis of NEUROD2, an effector transcription factor expressed in lineages of cortical projection neurons during the peak of cortical excitatory neurogenesis. Our results suggest that during cortical development NEUROD2 targets key genes that are required for Reelin signaling, a major pathway that regulates the migration of neurons from germinal zones to their final layers of residence within the cortex. We also find that NEUROD2 binds to a large set of genes with functions in layer-specific differentiation and in axonal pathfinding of cortical projection neurons. CONCLUSIONS: Our analysis of in vivo NEUROD2 target genes offers mechanistic insight into signaling pathways that regulate neuronal migration and axon guidance and identifies genes that are likely to be required for proper cortical development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Genome-Wide Association Study , Neurons/cytology , Neurons/metabolism , Neuropeptides/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Cell Lineage/genetics , Cerebral Cortex/embryology , Chromatin Immunoprecipitation , Gene Expression , High-Throughput Nucleotide Sequencing , Mice , Models, Biological , Neuropeptides/metabolism , Protein Binding , Reelin Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...