Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34983843

RESUMO

In Arabidopsis, vacuolar sorting receptor isoform 1 (VSR1) sorts 12S globulins to the protein storage vacuoles during seed development. Vacuolar sorting is mediated by specific protein-protein interactions between VSR1 and the vacuolar sorting determinant located at the C terminus (ctVSD) on the cargo proteins. Here, we determined the crystal structure of the protease-associated domain of VSR1 (VSR1-PA) in complex with the C-terminal pentapeptide (468RVAAA472) of cruciferin 1, an isoform of 12S globulins. The 468RVA470 motif forms a parallel ß-sheet with the switch III residues (127TMD129) of VSR1-PA, and the 471AA472 motif docks to a cradle formed by the cargo-binding loop (95RGDCYF100), making a hydrophobic interaction with Tyr99. The C-terminal carboxyl group of the ctVSD is recognized by forming salt bridges with Arg95. The C-terminal sequences of cruciferin 1 and vicilin-like storage protein 22 were sufficient to redirect the secretory red fluorescent protein (spRFP) to the vacuoles in Arabidopsis protoplasts. Adding a proline residue to the C terminus of the ctVSD and R95M substitution of VSR1 disrupted receptor-cargo interactions in vitro and led to increased secretion of spRFP in Arabidopsis protoplasts. How VSR1-PA recognizes ctVSDs of other storage proteins was modeled. The last three residues of ctVSD prefer hydrophobic residues because they form a hydrophobic cluster with Tyr99 of VSR1-PA. Due to charge-charge interactions, conserved acidic residues, Asp129 and Glu132, around the cargo-binding site should prefer basic residues over acidic ones in the ctVSD. The structural insights gained may be useful in targeting recombinant proteins to the protein storage vacuoles in seeds.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Substituição de Aminoácidos , Arabidopsis/química , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Cristalografia por Raios X , Mutação de Sentido Incorreto , Conformação Proteica em Folha beta , Domínios Proteicos , Transporte Proteico , Protoplastos/química , Protoplastos/metabolismo , Proteínas de Armazenamento de Sementes/química , Proteínas de Armazenamento de Sementes/genética , Proteínas de Armazenamento de Sementes/metabolismo , Relação Estrutura-Atividade , Vacúolos/química , Vacúolos/genética , Vacúolos/metabolismo
2.
J Hum Genet ; 64(4): 271-280, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30670789

RESUMO

A decade ago, we described novel de novo submicroscopic deletions of chromosome 14q11.2 in three children with developmental delay, cognitive impairment, and similar dysmorphic features, including widely-spaced eyes, short nose with flat nasal bridge, long philtrum, prominent Cupid's bow of the upper lip, full lower lip, and auricular anomalies. We suggested that this constituted a new multiple congenital anomaly-intellectual disability syndrome due to defects in CHD8 and/or SUPT16H. The three patients in our original cohort were between 2 years and 3 years of age at the time. Here we present a fourth patient and clinical updates on our previous patients. To document the longitudinal course more fully, we integrate published reports of other patients and describe genotype-phenotype correlations among them. Children with the disorder present with developmental delay, intellectual disability, and/or autism spectrum disorder in addition to characteristic facies. Gastrointestinal and sleep problems are notable. The identification of multiple patients with the same genetic defect and characteristic clinical phenotype, confirms our suggestion that this is a syndromic disorder caused by haploinsufficiency or heterozygous loss of function of CHD8.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Deficiência Intelectual/genética , Transtornos do Neurodesenvolvimento/genética , Fatores de Transcrição/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/fisiopatologia , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 14/genética , Fácies , Feminino , Haploinsuficiência/genética , Heterozigoto , Humanos , Deficiência Intelectual/fisiopatologia , Masculino , Megalencefalia/genética , Megalencefalia/fisiopatologia , Transtornos do Neurodesenvolvimento/patologia
3.
J Prosthodont ; 28(2): e722-e735, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29665177

RESUMO

PURPOSE: The purpose of this systematic review and meta-analysis was to review and analyze the prosthodontic complications, survival, and success of metal-ceramic (MC) and all-ceramic (AC) complete-arch fixed implant dental prostheses (CFIDPs) with a minimum mean follow-up period of 5 years. METHODS: A structured literature search was conducted using 3 electronic databases (MEDLINE, the Cochrane Library, Web of Science) for clinical studies reporting on prosthodontic complications of metal-ceramic and/or all-ceramic CFIDPs published between 2000 and 2016. This was complemented with hand searching in relevant journals, references, as well as searching in grey literature. Risk of bias analysis for randomized controlled trials was done following the recommendations from the Cochrane Collaboration. Quality appraisal for nonrandomized studies was executed according to the Newcastle-Ottawa scale (NOS). The final selection included only studies with a minimum mean follow-up time of 5 years. RESULTS: The electronic databases search yielded 1804 relevant titles and abstracts; 11 studies were finally selected (9 for MC and 2 for AC CFIDPs). Risk of bias in most selected studies was low. Heterogeneity across studies of MC CFIDPs was within acceptable range but not among AC CFIDP studies, so no meta-analysis was performed for the latter. Regarding MC CFIDPs, most studies recorded 100% survival rate (survival range: 92.4-100%, success range: 47-96.7%), with veneer fracture being the most-common complication. Five- and 10-year cumulative complication rates for MC CFIDP veneer fractures were 22.1% and 39.3%, respectively, but with variable confidence intervals. The 2 studies included for AC CFIDPs reported 100% survival rates but differed in success rates, with the one using predominantly monolithic zirconia restorations reporting 90.9%, and the one using bi-layered zirconia reporting 60.4%, with complications attributed to veneer fracture. CONCLUSIONS: MC and AC CFIDPs presented with veneer fractures as primary complication. This may require significant maintenance. Other complications were negligible after a mean follow-up period of at least 5 years. More long-term studies, especially on all-ceramic CFIDPs are needed.


Assuntos
Cerâmica , Prótese Dentária Fixada por Implante , Prótese Total , Ligas Metalo-Cerâmicas , Planejamento de Prótese Dentária , Falha de Restauração Dentária , Seguimentos , Humanos , Fatores de Tempo
4.
J Lipid Res ; 56(2): 358-68, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25510249

RESUMO

Inflammation of adipose tissue induces metabolic derangements associated with obesity. Thus, determining ways to control or inhibit inflammation in adipose tissue is of clinical interest. The present study tested the hypothesis that in mouse adipose tissue, endogenous prostaglandin E2 (PGE2) negatively regulates inflammation via activation of prostaglandin E receptor 4 (EP4). PGE2 (5-500 nM) attenuated lipopolysaccharide-induced mRNA and protein expression of chemokines, including interferon-γ-inducible protein 10 and macrophage-inflammatory protein-1α in mouse adipose tissue. A selective EP4 antagonist (L161,982) reversed, and two structurally different selective EP4 agonists [CAY10580 and CAY10598] mimicked these actions of PGE2. Adipose tissue derived from EP4-deficient mice did not display this response. These findings establish the involvement of EP4 receptors in this anti-inflammatory response. Experiments performed on adipose tissue from high-fat-fed mice demonstrated EP4-dependent attenuation of chemokine production during diet-induced obesity. The anti-inflammatory actions of EP4 became more important on a high-fat diet, in that EP4 activation suppressed a greater variety of chemokines. Furthermore, adipose tissue and systemic inflammation was enhanced in high-fat-fed EP4-deficient mice compared with wild-type littermates, and in high-fat-fed untreated C57BL/6 mice compared with mice treated with EP4 agonist. These findings provide in vivo evidence that PGE2-EP4 signaling limits inflammation. In conclusion, PGE2, via activation of EP4 receptors, functions as an endogenous anti-inflammatory mediator in mouse adipose tissue, and targeting EP4 may mitigate adipose tissue inflammation.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Quimiocinas/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Animais , Quimiocina CCL3/metabolismo , Quimiocina CXCL10/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/farmacologia , Técnicas In Vitro , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Interleucina-8/farmacologia , Masculino , Pirrolidinonas/farmacologia , Receptores de Prostaglandina E Subtipo EP4/agonistas , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Proteína Amiloide A Sérica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tetrazóis/farmacologia , Tiofenos/farmacologia , Triazóis/farmacologia
5.
DNA Cell Biol ; 43(6): 267-270, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38579130

RESUMO

Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have shown efficacy in the treatment of metabolic disease-related complications, partially attributable to their anti-inflammatory properties. However, the specific cell types and pathways involved in these effects were not fully understood. A recent study by Wong et al. demonstrated the importance of the brain GLP-1R in mediating the anti-inflammatory effects of GLP-1RAs in Toll-like receptor and sepsis-mediated inflammation. In this discussion, we review the existing literature on the action of GLP-1RAs in inflammation and explore the implications of these recent findings.


Assuntos
Anti-Inflamatórios , Receptor do Peptídeo Semelhante ao Glucagon 1 , Inflamação , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Humanos , Anti-Inflamatórios/farmacologia , Inflamação/tratamento farmacológico , Animais , Neuroimunomodulação/efeitos dos fármacos , Receptores Toll-Like/agonistas , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Agonistas do Receptor do Peptídeo 1 Semelhante ao Glucagon
6.
Cell Metab ; 36(1): 130-143.e5, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38113888

RESUMO

Glucagon-like peptide-1 receptor agonists (GLP-1RAs) exert anti-inflammatory effects relevant to the chronic complications of type 2 diabetes. Although GLP-1RAs attenuate T cell-mediated gut and systemic inflammation directly through the gut intraepithelial lymphocyte GLP-1R, how GLP-1RAs inhibit systemic inflammation in the absence of widespread immune expression of the GLP-1R remains uncertain. Here, we show that GLP-1R activation attenuates the induction of plasma tumor necrosis factor alpha (TNF-α) by multiple Toll-like receptor agonists. These actions are not mediated by hematopoietic or endothelial GLP-1Rs but require central neuronal GLP-1Rs. In a cecal slurry model of polymicrobial sepsis, GLP-1RAs similarly require neuronal GLP-1Rs to attenuate detrimental responses associated with sepsis, including sickness, hypothermia, systemic inflammation, and lung injury. Mechanistically, GLP-1R activation leads to reduced TNF-α via α1-adrenergic, δ-opioid, and κ-opioid receptor signaling. These data extend emerging concepts of brain-immune networks and posit a new gut-brain GLP-1R axis for suppression of peripheral inflammation.


Assuntos
Diabetes Mellitus Tipo 2 , Sepse , Humanos , Exenatida , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeos/farmacologia , Agonistas do Receptor Semelhante a Toll , Peçonhas/farmacologia , Fator de Necrose Tumoral alfa , Inflamação , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo
7.
J Infect Dis ; 206(3): 341-51, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22615319

RESUMO

A scarlet fever outbreak occurred in Hong Kong in 2011. The majority of cases resulted in the isolation of Streptococcus pyogenes emm12 with multiple antibiotic resistances. Phylogenetic analysis of 22 emm12 scarlet fever outbreak isolates, 7 temporally and geographically matched emm12 non-scarlet fever isolates, and 18 emm12 strains isolated during 2005-2010 indicated the outbreak was multiclonal. Genome sequencing of 2 nonclonal scarlet fever isolates (HKU16 and HKU30), coupled with diagnostic polymerase chain reaction assays, identified 2 mobile genetic elements distributed across the major lineages: a 64.9-kb integrative and conjugative element encoding tetracycline and macrolide resistance and a 46.4-kb prophage encoding superantigens SSA and SpeC and the DNase Spd1. Phenotypic comparison of HKU16 and HKU30 with the S. pyogenes M1T1 strain 5448 revealed that HKU16 displays increased adherence to HEp-2 human epithelial cells, whereas HKU16, HKU30, and 5448 exhibit equivalent resistance to neutrophils and virulence in a humanized plasminogen murine model. However, in contrast to M1T1, the virulence of HKU16 and HKU30 was not associated with covRS mutation. The multiclonal nature of the emm12 scarlet fever isolates suggests that factors such as mobile genetic elements, environmental factors, and host immune status may have contributed to the 2011 scarlet fever outbreak.


Assuntos
Surtos de Doenças , Escarlatina/epidemiologia , Escarlatina/microbiologia , Streptococcus pyogenes/classificação , Streptococcus pyogenes/genética , Adolescente , Adulto , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/fisiologia , Genoma Bacteriano , Genômica , Hong Kong/epidemiologia , Humanos , Lactente , Sequências Repetitivas Dispersas , Masculino , Pessoa de Meia-Idade , Epidemiologia Molecular , Fenótipo , Filogenia , Streptococcus pyogenes/efeitos dos fármacos
8.
Dev Cell ; 58(6): 489-505.e7, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36898377

RESUMO

Loss of muscle mass is a common manifestation of chronic disease. We find the canonical Wnt pathway to be activated in mesenchymal progenitors (MPs) from cancer-induced cachectic mouse muscle. Next, we induce ß-catenin transcriptional activity in murine MPs. As a result, we observe expansion of MPs in the absence of tissue damage, as well as rapid loss of muscle mass. Because MPs are present throughout the organism, we use spatially restricted CRE activation and show that the induction of tissue-resident MP activation is sufficient to induce muscle atrophy. We further identify increased expression of stromal NOGGIN and ACTIVIN-A as key drivers of atrophic processes in myofibers, and we verify their expression by MPs in cachectic muscle. Finally, we show that blocking ACTIVIN-A rescues the mass loss phenotype triggered by ß-catenin activation in MPs, confirming its key functional role and strengthening the rationale for targeting this pathway in chronic disease.


Assuntos
Via de Sinalização Wnt , beta Catenina , Camundongos , Animais , beta Catenina/metabolismo , Ativinas , Músculos/metabolismo
9.
Mol Metab ; 66: 101641, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36396031

RESUMO

OBJECTIVES: Glucagon-like peptide-1 receptor (GLP-1R) agonists reduce the rates of major cardiovascular events, including myocardial infarction in people with type 2 diabetes, and decrease infarct size while preserving ventricular function in preclinical studies. Nevertheless, the precise cellular sites of GLP-1R expression that mediate the cardioprotective actions of GLP-1 in the setting of ischemic cardiac injury are uncertain. METHODS: Publicly available single cell RNA sequencing (scRNA-seq) datasets on mouse and human heart cells were analyzed for Glp1r/GLP1R expression. Fluorescent activated cell sorting was used to localize Glp1r expression in cell populations from the mouse heart. The importance of endothelial and hematopoietic cells for the cardioprotective response to liraglutide in the setting of acute myocardial infarction (MI) was determined by inactivating the Glp1r in Tie2+ cell populations. Cardiac gene expression profiles regulated by liraglutide were examined using RNA-seq to interrogate mouse atria and both infarcted and non-infarcted ventricular tissue after acute coronary artery ligation. RESULTS: In mice, cardiac Glp1r mRNA transcripts were exclusively detected in endocardial cells by scRNA-seq. In contrast, analysis of human heart by scRNA-seq localized GLP1R mRNA transcripts to populations of atrial and ventricular cardiomyocytes. Moreover, very low levels of GIPR, GCGR and GLP2R mRNA transcripts were detected in the human heart. Cell sorting and RNA analyses detected cardiac Glp1r expression in endothelial cells (ECs) within the atria and ventricle in the ischemic and non-ischemic mouse heart. Transcriptional responses to liraglutide administration were not evident in wild type mouse ventricles following acute MI, however liraglutide differentially regulated genes important for inflammation, cardiac repair, cell proliferation, and angiogenesis in the left atrium, while reducing circulating levels of IL-6 and KC/GRO within hours of acute MI. Inactivation of the Glp1r within the Tie2+ cell expression domain encompassing ECs revealed normal cardiac structure and function, glucose homeostasis and body weight in Glp1rTie2-/- mice. Nevertheless, the cardioprotective actions of liraglutide to reduce infarct size, augment ejection fraction, and improve survival after experimental myocardial infarction (MI), were attenuated in Glp1rTie2-/- mice. CONCLUSIONS: These findings identify the importance of the murine Tie2+ endothelial cell GLP-1R as a target for the cardioprotective actions of GLP-1R agonists and support the importance of the atrial and ventricular endocardial GLP-1R as key sites of GLP-1 action in the ischemic mouse heart. Hitherto unexplored species-specific differences in cardiac GLP-1R expression challenge the exclusive use of mouse models for understanding the mechanisms of GLP-1 action in the normal and ischemic human heart.


Assuntos
Fibrilação Atrial , Receptor do Peptídeo Semelhante ao Glucagon 1 , Liraglutida , Infarto do Miocárdio , Animais , Humanos , Camundongos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Células Endoteliais/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Liraglutida/farmacologia , Infarto do Miocárdio/tratamento farmacológico , RNA Mensageiro , Modelos Animais de Doenças , Receptor TIE-2/metabolismo
10.
Cell Metab ; 34(10): 1514-1531.e7, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36027914

RESUMO

Gut intraepithelial lymphocytes (IELs) are thought to calibrate glucagon-like peptide 1 (GLP-1) bioavailability, thereby regulating systemic glucose and lipid metabolism. Here, we show that the gut IEL GLP-1 receptor (GLP-1R) is not required for enteroendocrine L cell GLP-1 secretion and glucose homeostasis nor for the metabolic benefits of GLP-1R agonists (GLP-1RAs). Instead, the gut IEL GLP-1R is essential for the full effects of GLP-1RAs on gut microbiota. Moreover, independent of glucose control or weight loss, the anti-inflammatory actions of GLP-1RAs require the gut IEL GLP-1R to selectively restrain local and systemic T cell-induced, but not lipopolysaccharide-induced, inflammation. Such effects are mediated by the suppression of gut IEL effector functions linked to the dampening of proximal T cell receptor signaling in a protein-kinase-A-dependent manner. These data reposition key roles of the L cell-gut IEL GLP-1R axis, revealing mechanisms linking GLP-1R activation in gut IELs to modulation of microbiota composition and control of intestinal and systemic inflammation.


Assuntos
Microbioma Gastrointestinal , Linfócitos Intraepiteliais , Glicemia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/metabolismo , Humanos , Inflamação , Intestinos , Linfócitos Intraepiteliais/metabolismo , Receptores de Antígenos de Linfócitos T
11.
Diabetes ; 71(5): 1115-1127, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35192688

RESUMO

The incretin hormone glucose-dependent insulinotropic polypeptide (GIP) augments glucose-dependent insulin secretion through its receptor expressed on islet ß-cells. GIP also acts on adipose tissue; yet paradoxically, both enhanced and reduced GIP receptor (GIPR) signaling reduce adipose tissue mass and attenuate weight gain in response to nutrient excess. Moreover, the precise cellular localization of GIPR expression within white adipose tissue (WAT) remains uncertain. We used mouse genetics to target Gipr expression within adipocytes. Surprisingly, targeting Cre expression to adipocytes using the adiponectin (Adipoq) promoter did not produce meaningful reduction of WAT Gipr expression in Adipoq-Cre:Giprflx/flx mice. In contrast, adenoviral expression of Cre under the control of the cytomegalovirus promoter, or transgenic expression of Cre using nonadipocyte-selective promoters (Ap2/Fabp4 and Ubc) markedly attenuated WAT Gipr expression. Analysis of single-nucleus RNA-sequencing, adipose tissue data sets localized Gipr/GIPR expression predominantly to pericytes and mesothelial cells rather than to adipocytes. Together, these observations reveal that adipocytes are not the major GIPR+ cell type within WAT-findings with mechanistic implications for understanding how GIP and GIP-based co-agonists control adipose tissue biology.


Assuntos
Receptores dos Hormônios Gastrointestinais , Tecido Adiposo Branco/metabolismo , Animais , Polipeptídeo Inibidor Gástrico/metabolismo , Glucose , Camundongos , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo
12.
JCI Insight ; 6(22)2021 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-34673572

RESUMO

Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are used to treat diabetes and obesity and reduce rates of major cardiovascular events, such as stroke and myocardial infarction. Nevertheless, the identity of GLP-1R-expressing cell types mediating the cardiovascular benefits of GLP-1RA remains incompletely characterized. Herein, we investigated the importance of murine Glp1r expression within endothelial and hematopoietic cells. Mice with targeted inactivation of Glp1r in Tie2+ cells exhibited reduced levels of Glp1r mRNA transcripts in aorta, liver, spleen, blood, and gut. Glp1r expression in bone marrow cells was very low and not further reduced in Glp1rTie2-/- mice. The GLP-1RA semaglutide reduced the development of atherosclerosis induced by viral PCSK9 expression in both Glp1rTie2+/+ and Glp1rTie2-/- mice. Hepatic Glp1r mRNA transcripts were reduced in Glp1rTie2-/- mice, and liver Glp1r expression was localized to γδ T cells. Moreover, semaglutide reduced hepatic Tnf, Abcg1, Tgfb1, Cd3g, Ccl2, and Il2 expression; triglyceride content; and collagen accumulation in high-fat, high-cholesterol diet-fed Glp1rTie2+/+ mice but not Glp1rTie2-/- mice. Collectively, these findings demonstrate that Tie2+ endothelial or hematopoietic cell GLP-1Rs are dispensable for the antiatherogenic actions of GLP-1RA, whereas Tie2-targeted GLP-1R+ cells are required for a subset of the antiinflammatory actions of semaglutide in the liver.


Assuntos
Aterosclerose/tratamento farmacológico , Células Endoteliais/metabolismo , Peptídeos Semelhantes ao Glucagon/uso terapêutico , Células-Tronco Hematopoéticas/metabolismo , Fígado/efeitos dos fármacos , Síndrome Metabólica/tratamento farmacológico , Animais , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeos Semelhantes ao Glucagon/farmacologia , Humanos , Masculino , Camundongos
13.
Endocr Rev ; 42(2): 101-132, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33320179

RESUMO

Glucagon-like peptide-1 (GLP-1) is produced in gut endocrine cells and in the brain, and acts through hormonal and neural pathways to regulate islet function, satiety, and gut motility, supporting development of GLP-1 receptor (GLP-1R) agonists for the treatment of diabetes and obesity. Classic notions of GLP-1 acting as a meal-stimulated hormone from the distal gut are challenged by data supporting production of GLP-1 in the endocrine pancreas, and by the importance of brain-derived GLP-1 in the control of neural activity. Moreover, attribution of direct vs indirect actions of GLP-1 is difficult, as many tissue and cellular targets of GLP-1 action do not exhibit robust or detectable GLP-1R expression. Furthermore, reliable detection of the GLP-1R is technically challenging, highly method dependent, and subject to misinterpretation. Here we revisit the actions of GLP-1, scrutinizing key concepts supporting gut vs extra-intestinal GLP-1 synthesis and secretion. We discuss new insights refining cellular localization of GLP-1R expression and integrate recent data to refine our understanding of how and where GLP-1 acts to control inflammation, cardiovascular function, islet hormone secretion, gastric emptying, appetite, and body weight. These findings update our knowledge of cell types and mechanisms linking endogenous vs pharmacological GLP-1 action to activation of the canonical GLP-1R, and the control of metabolic activity in multiple organs.


Assuntos
Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Humanos , Obesidade
14.
Mol Metab ; 48: 101213, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33741532

RESUMO

OBJECTIVE: Glucose-dependent insulinotropic polypeptide (GIP) and Glucagon-like peptide-1 (GLP-1) are incretin hormones that exert overlapping yet distinct actions on islet ß-cells. We recently observed that GIP, but not GLP-1, upregulated islet expression of Transcription Factor 7 (TCF7), a gene expressed in immune cells and associated with the risk of developing type 1 diabetes. TCF7 has also been associated with glucose homeostasis control in the liver. Herein we studied the relative metabolic importance of TCF7 expression in hepatocytes vs. islet ß-cells in mice. METHODS: Tcf7 expression was selectively inactivated in adult mouse hepatocytes using adenoviral Cre expression and targeted in ß-cells using two different lines of insulin promoter-Cre mice. Glucose homeostasis, plasma insulin and triglyceride responses, islet histology, hepatic and islet gene expression, and body weight gain were evaluated in mice fed regular chow or high fat diets. Tcf7 expression within pancreatic islets and immune cells was evaluated using published single cell RNA-seq (scRNA-seq) data, and in islet RNA from immunodeficient Rag2-/-Il2rg-/- mice. RESULTS: Reduction of hepatocyte Tcf7 expression did not impair glucose homeostasis, lipid tolerance or hepatic gene expression profiles linked to control of metabolic or immune pathways. Similarly, oral and intraperitoneal glucose tolerance, plasma insulin responses, islet histology, body weight gain, and insulin tolerance were not different in mice with targeted recombination of Tcf7 in insulin-positive ß-cells. Surprisingly, islet Tcf7 mRNA transcripts were not reduced in total islet RNA containing endocrine and associated non-endocrine cell types from Tcf7ßcell-/- mice, despite Cre-mediated recombination of islet genomic DNA. Furthermore, glucose tolerance was normal in whole body Tcf7-/- mice. Analysis of scRNA-seq datasets localized pancreatic Tcf7 expression to islet progenitors during development, and immune cells, but not within differentiated islet ß-cells or endocrine lineages within mature islets. Moreover, the expression of Tcf7 was extremely low in islet RNA from Rag2-/-Il2rg-/- mice and, consistent with expression within immune cells, Tcf7 was highly correlated with levels of Cd3g mRNA transcripts in RNA from wild type mouse islets. CONCLUSIONS: These findings demonstrate that Tcf7 expression is not a critical determinant of glucose homeostasis in mice. Moreover, the detection of Tcf7 expression within islet mRNA is attributable to the expression of Tcf7 RNA in islet-associated murine immune cells, and not in islet ß-cells.


Assuntos
Glicemia/metabolismo , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Homeostase/genética , Células Secretoras de Insulina/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/genética , Insulina/sangue , Insulina/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Triglicerídeos/sangue , Aumento de Peso/genética
15.
J Phys Chem A ; 114(14): 4762-9, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20095599

RESUMO

Infrared vibrational predissociation spectra are recorded for Br(-)-(NH(3))(n) (n = 1-4) and I(-)-NH(3) clusters in the N-H stretch region (3040-3460 cm(-1)). To aid spectral assignments and clarify structures of the Br(-)-(NH(3))(n) clusters, ab initio calculations are performed at the MP2/aug-cc-pVDZ and MP2/aug-cc-pVTZ levels of theory. The Br(-)-NH(3) and I(-)-NH(3) dimers are predicted to have structures in which the NH(3) molecule is attached to the halide anion by a single hydrogen-bond. The dominant infrared band for Br(-)-NH(3) at 3171 cm(-1) corresponds to a hydrogen-bonded N-H stretch vibrational mode, whereas two weaker bands are assigned to a symmetric stretch vibration of the nonbonded N-H groups (3347 cm(-1)) and to an ammonia-based bending overtone (3293 cm(-1)) deriving infrared intensity through Fermi interaction with the H-bonded N-H stretch mode. The corresponding I(-)-NH(3) spectrum is dominated by the H-bonded N-H stretch band at 3217 cm(-1), with three weaker bands at 3240, 3305, and 3360 cm(-1) assigned to two bending overtone vibrations and the nonbonded N-H symmetric stretch vibration, respectively. Spectra of the Br(-)-(NH(3))(n), n = 2-4, clusters are similar to the I(-)-NH(3) spectrum, exhibiting evidence for strong Fermi interactions between the H-bonded N-H stretch vibrational mode and ammonia-based bending overtones. On the basis of the infrared spectra and ab initio calculations, the larger Br(-)-(NH(3))(n) clusters are deduced to have structures in which the NH(3) molecules are attached to the Br(-) by single H-bonds, but not necessarily to one other.


Assuntos
Amônia/química , Bromo/química , Iodo/química , Dimerização , Modelos Moleculares , Conformação Molecular , Espectrofotometria Infravermelho
16.
Cureus ; 12(11): e11571, 2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33282601

RESUMO

In 2019, we confirmed that the haploinsufficiency of CHD8 does indeed cause the novel syndromic neurodevelopmental disease we first discovered a dozen years before. Here, we report the first whole transcriptome RNAseq gene expression profiling for a patient with this new syndrome, as a preliminary exploration of potential pathophysiological mechanisms. We compared our patient transcriptome profile with that of all publicly available RNAseq datasets from human cellular models including neuronal progenitor cells, neurons and organoids. We compared differential gene expression profiles overall and conducted phenotype-informed data filtration based on the characteristic syndrome presentation. We found that concordance among differential gene expression profiles was poor across all datasets. Nevertheless, remarkably, we show that the patient blood differential gene expression profile most resembled that of the neuronal cell model, a finding that encourages further transcriptome profiling using patient blood samples. In addition, our custom phenotype-informed analyses yielded important, differentially expressed syndrome pathophysiology target genes. Finally, we note that genes dysregulated due to CHD8 heterozygous deletion are linked to known neurological as well as oncological pathways.

17.
J Exp Med ; 217(8)2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32478834

RESUMO

Adipose tissue (AT) regulatory T cells (T regs) control inflammation and metabolism. Diet-induced obesity causes hyperinsulinemia and diminishes visceral AT (VAT) T reg number and function, but whether these two phenomena were mechanistically linked was unknown. Using a T reg-specific insulin receptor (Insr) deletion model, we found that diet-induced T reg dysfunction is driven by T reg-intrinsic insulin signaling. Compared with Foxp3cre mice, after 13 wk of high-fat diet, Foxp3creInsrfl/fl mice exhibited improved glucose tolerance and insulin sensitivity, effects associated with lower AT inflammation and increased numbers of ST2+ T regs in brown AT, but not VAT. Similarly, Foxp3creInsrfl/fl mice were protected from the metabolic effects of aging, but surprisingly had reduced VAT T regs and increased VAT inflammation compared with Foxp3cre mice. Thus, in both diet- and aging-associated hyperinsulinemia, excessive Insr signaling in T regs leads to undesirable metabolic outcomes. Ablation of Insr signaling in T regs represents a novel approach to mitigate the detrimental effects of hyperinsulinemia on immunoregulation of metabolic syndrome.


Assuntos
Envelhecimento/imunologia , Dieta Hiperlipídica/efeitos adversos , Gordura Intra-Abdominal/imunologia , Síndrome Metabólica/imunologia , Receptor de Insulina/deficiência , Linfócitos T Reguladores/imunologia , Envelhecimento/genética , Envelhecimento/patologia , Animais , Deleção de Genes , Gordura Intra-Abdominal/patologia , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Camundongos , Camundongos Transgênicos , Receptor de Insulina/imunologia , Linfócitos T Reguladores/patologia
18.
Diabetes ; 67(3): 412-422, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29217654

RESUMO

p300 (EP300) and CBP (CREBBP) are transcriptional coactivators with histone acetyltransferase activity. Various ß-cell transcription factors can recruit p300/CBP, and thus the coactivators could be important for ß-cell function and health in vivo. We hypothesized that p300/CBP contribute to the development and proper function of pancreatic islets. To test this, we bred and studied mice lacking p300/CBP in their islets. Mice lacking either p300 or CBP in islets developed glucose intolerance attributable to impaired insulin secretion, together with reduced α- and ß-cell area and islet insulin content. These phenotypes were exacerbated in mice with only a single copy of p300 or CBP expressed in islets. Removing p300 in pancreatic endocrine progenitors impaired proliferation of neonatal α- and ß-cells. Mice lacking all four copies of p300/CBP in pancreatic endocrine progenitors failed to establish α- and ß-cell mass postnatally. Transcriptomic analyses revealed significant overlaps between p300/CBP-downregulated genes and genes downregulated in Hnf1α-null islets and Nkx2.2-null islets, among others. Furthermore, p300/CBP are important for the acetylation of H3K27 at loci downregulated in Hnf1α-null islets. We conclude that p300 and CBP are limiting cofactors for islet development, and hence for postnatal glucose homeostasis, with some functional redundancy.


Assuntos
Proteína de Ligação a CREB/metabolismo , Proliferação de Células , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Células-Tronco/metabolismo , Acetilação , Animais , Animais Recém-Nascidos , Glicemia/análise , Proteína de Ligação a CREB/genética , Tamanho Celular , Cruzamentos Genéticos , Proteína p300 Associada a E1A/genética , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/patologia , Intolerância à Glucose/sangue , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Histonas/metabolismo , Proteína Homeobox Nkx-2.2 , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Lisina , Camundongos Knockout , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Células-Tronco/citologia , Células-Tronco/patologia
19.
Cell Rep ; 22(1): 163-174, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29298418

RESUMO

Depolarization of neuroendocrine cells results in calcium influx, which induces vesicle exocytosis and alters gene expression. These processes, along with the restoration of resting membrane potential, are energy intensive. We hypothesized that cellular mechanisms exist to maximize energy production during excitation. Here, we demonstrate that NPAS4, an immediate early basic helix-loop-helix (bHLH)-PAS transcription factor, acts to maximize energy production by suppressing hypoxia-inducible factor 1α (HIF1α). As such, knockout of Npas4 from insulin-producing ß cells results in reduced OXPHOS, loss of insulin secretion, ß cell dedifferentiation, and type 2 diabetes. NPAS4 plays a similar role in the nutrient-sensing cells of the hypothalamus. Its knockout here results in increased food intake, reduced locomotor activity, and elevated peripheral glucose production. In conclusion, NPAS4 is critical for the coordination of metabolism during the stimulation of electrically excitable cells; its loss leads to the defects in cellular metabolism that underlie the cellular dysfunction that occurs in metabolic disease.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipotálamo/metabolismo , Células Neuroendócrinas/metabolismo , Fosforilação Oxidativa , Oxigênio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipotálamo/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/citologia
20.
PLoS One ; 12(3): e0174030, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28301585

RESUMO

Maternal overnutrition and obesity during pregnancy can have long-term effects on offspring physiology and behaviour. These developmental programming effects may be mediated by fetal exposure to glucocorticoids, which is regulated in part by placental 11ß-hydroxysteroid dehydrogenase (11ß-HSD) type 1 and 2. We tested whether a maternal high-fat, high-sucrose diet would alter expression of placental 11ß-HSD1 and 2, thereby increasing fetal exposure to maternal glucocorticoids, with downstream effects on offspring physiology and behaviour. C57BL/6J mice were fed a high-fat, high-sucrose (HFHS) diet or a nutrient-matched low-fat, no-sucrose control diet prior to and during pregnancy and lactation. At day 17 of gestation, HFHS dams had ~20% lower circulating corticosterone levels than controls. Furthermore, there was a significant interaction between maternal diet and fetal sex for circulating corticosterone levels in the fetuses, whereby HFHS males tended to have higher corticosterone than control males, with no effect in female fetuses. However, placental 11ß-HSD1 or 11ß-HSD2 expression did not differ between diets or show an interaction between diet and sex. To assess potential long-term consequences of this sex-specific effect on fetal corticosterone, we studied locomotor activity and metabolic traits in adult offspring. Despite a sex-specific effect of maternal diet on fetal glucocorticoids, there was little evidence of sex-specific effects on offspring physiology or behaviour, although HFHS offspring of both sexes had higher circulating corticosterone at 9 weeks of age. Our results suggest the existence of as yet unknown mechanisms that mitigate the effects of altered glucocorticoid exposure early in development, making offspring resilient to the potentially negative effects of a HFHS maternal diet.


Assuntos
Dieta Hiperlipídica , Sacarose Alimentar/administração & dosagem , Feto/metabolismo , Glucocorticoides/metabolismo , Atividade Motora , Fatores Sexuais , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa