Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Endocrinol ; 261(3)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593829

RESUMO

Pancreatic alpha cell activity and glucagon secretion lower as glucose levels increase. While part of the decrease is regulated by glucose itself, paracrine signaling by their neighboring beta and delta cells also plays an important role. Somatostatin from delta cells is an important local inhibitor of alpha cells at high glucose. Additionally, urocortin 3 (UCN3) is a hormone that is co-released from beta cells with insulin and acts locally to potentiate somatostatin secretion from delta cells. UCN3 thus inhibits insulin secretion via a negative feedback loop with delta cells, but its role with respect to alpha cells and glucagon secretion is not understood. We hypothesize that the somatostatin-driven glucagon inhibition at high glucose is regulated in part by UCN3 from beta cells. Here, we use a combination of live functional Ca2+ and cAMP imaging as well as direct glucagon secretion measurement, all from alpha cells in intact mouse islets, to determine the contributions of UCN3 to alpha cell behavior. Exogenous UCN3 treatment decreased alpha cell Ca2+ and cAMP levels and inhibited glucagon release. Blocking endogenous UCN3 signaling increased alpha cell Ca2+ by 26.8 ± 7.6%, but this did not result in increased glucagon release at high glucose. Furthermore, constitutive deletion of Ucn3 did not increase Ca2+ activity or glucagon secretion relative to controls. UCN3 is thus capable of inhibiting mouse alpha cells, but, given the subtle effects of endogenous UCN3 signaling on alpha cells, we propose that UCN3-driven somatostatin may serve to regulate local paracrine glucagon levels in the islet instead of inhibiting gross systemic glucagon release.


Assuntos
Células Secretoras de Glucagon , Glucagon , Comunicação Parácrina , Urocortinas , Animais , Urocortinas/metabolismo , Urocortinas/genética , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Camundongos , Glucagon/metabolismo , Glucose/metabolismo , Cálcio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , AMP Cíclico/metabolismo , Somatostatina/farmacologia , Somatostatina/metabolismo
2.
Biochemistry ; 62(21): 3050-3060, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37813856

RESUMO

Over the past decade, advances in genomics have identified thousands of additional protein-coding small open reading frames (smORFs) missed by traditional gene finding approaches. These smORFs encode peptides and small proteins, commonly termed micropeptides or microproteins. Several of these newly discovered microproteins have biological functions and operate through interactions with proteins and protein complexes within the cell. CYREN1 is a characterized microprotein that regulates double-strand break repair in mammalian cells through interaction with Ku70/80 heterodimer. Ku70/80 binds to and stabilizes double-strand breaks and recruits the machinery needed for nonhomologous end join repair. In this study, we examined the biochemical properties of CYREN1 to better understand and explain its cellular protein interactions. Our findings support that CYREN1 is an intrinsically disordered microprotein and this disordered structure allows it to enriches several proteins, including a newly discovered interaction with SF3B1 via a distinct short linear motif (SLiMs) on CYREN1. Since many microproteins are predicted to be disordered, CYREN1 is an exemplar of how microproteins interact with other proteins and reveals an unknown scaffolding function of this microprotein that may link NHEJ and splicing.


Assuntos
Peptídeos , Proteínas , Animais , Proteínas/genética , Peptídeos/genética , Fases de Leitura Aberta , Mamíferos/genética , Micropeptídeos
3.
Nat Commun ; 14(1): 1328, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36899004

RESUMO

The TINCR (Terminal differentiation-Induced Non-Coding RNA) gene is selectively expressed in epithelium tissues and is involved in the control of human epidermal differentiation and wound healing. Despite its initial report as a long non-coding RNA, the TINCR locus codes for a highly conserved ubiquitin-like microprotein associated with keratinocyte differentiation. Here we report the identification of TINCR as a tumor suppressor in squamous cell carcinoma (SCC). TINCR is upregulated by UV-induced DNA damage in a TP53-dependent manner in human keratinocytes. Decreased TINCR protein expression is prevalently found in skin and head and neck squamous cell tumors and TINCR expression suppresses the growth of SCC cells in vitro and in vivo. Consistently, Tincr knockout mice show accelerated tumor development following UVB skin carcinogenesis and increased penetrance of invasive SCCs. Finally, genetic analyses identify loss-of-function mutations and deletions encompassing the TINCR gene in SCC clinical samples supporting a tumor suppressor role in human cancer. Altogether, these results demonstrate a role for TINCR as protein coding tumor suppressor gene recurrently lost in squamous cell carcinomas.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , RNA Longo não Codificante , Animais , Camundongos , Humanos , Ubiquitina/metabolismo , Carcinoma de Células Escamosas/genética , Genes Supressores de Tumor , Queratinócitos/metabolismo , Neoplasias de Cabeça e Pescoço/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Micropeptídeos
4.
Cell Metab ; 35(1): 166-183.e11, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36599300

RESUMO

Microproteins (MPs) are a potentially rich source of uncharacterized metabolic regulators. Here, we use ribosome profiling (Ribo-seq) to curate 3,877 unannotated MP-encoding small ORFs (smORFs) in primary brown, white, and beige mouse adipocytes. Of these, we validated 85 MPs by proteomics, including 33 circulating MPs in mouse plasma. Analyses of MP-encoding mRNAs under different physiological conditions (high-fat diet) revealed that numerous MPs are regulated in adipose tissue in vivo and are co-expressed with established metabolic genes. Furthermore, Ribo-seq provided evidence for the translation of Gm8773, which encodes a secreted MP that is homologous to human and chicken FAM237B. Gm8773 is highly expressed in the arcuate nucleus of the hypothalamus, and intracerebroventricular administration of recombinant mFAM237B showed orexigenic activity in obese mice. Together, these data highlight the value of this adipocyte MP database in identifying MPs with roles in fundamental metabolic and physiological processes such as feeding.


Assuntos
Adipócitos Brancos , Tecido Adiposo Marrom , Humanos , Animais , Camundongos , Adipócitos Brancos/metabolismo , Tecido Adiposo Marrom/metabolismo , Fases de Leitura Aberta/genética , Tecido Adiposo Branco/metabolismo , Adipócitos Marrons/metabolismo , Micropeptídeos
5.
Nat Chem Biol ; 19(2): 187-197, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36266352

RESUMO

Lipids contribute to the structure, development, and function of healthy brains. Dysregulated lipid metabolism is linked to aging and diseased brains. However, our understanding of lipid metabolism in aging brains remains limited. Here we examined the brain lipidome of mice across their lifespan using untargeted lipidomics. Co-expression network analysis highlighted a progressive decrease in 3-sulfogalactosyl diacylglycerols (SGDGs) and SGDG pathway members, including the potential degradation products lyso-SGDGs. SGDGs show an age-related decline specifically in the central nervous system and are associated with myelination. We also found that an SGDG dramatically suppresses LPS-induced gene expression and release of pro-inflammatory cytokines from macrophages and microglia by acting on the NF-κB pathway. The detection of SGDGs in human and macaque brains establishes their evolutionary conservation. This work enhances interest in SGDGs regarding their roles in aging and inflammatory diseases and highlights the complexity of the brain lipidome and potential biological functions in aging.


Assuntos
Envelhecimento , Lipídeos , Animais , Humanos , Camundongos , Envelhecimento/genética , Anti-Inflamatórios , Encéfalo/metabolismo , Microglia/metabolismo , NF-kappa B/metabolismo
6.
J Biol Chem ; 295(18): 5891-5905, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32152231

RESUMO

Fatty acid esters of hydroxy fatty acids (FAHFAs) are a newly discovered class of signaling lipids with anti-inflammatory and anti-diabetic properties. However, the endogenous regulation of FAHFAs remains a pressing but unanswered question. Here, using MS-based FAHFA hydrolysis assays, LC-MS-based lipidomics analyses, and activity-based protein profiling, we found that androgen-induced gene 1 (AIG1) and androgen-dependent TFPI-regulating protein (ADTRP), two threonine hydrolases, control FAHFA levels in vivo in both genetic and pharmacologic mouse models. Tissues from mice lacking ADTRP (Adtrp-KO), or both AIG1 and ADTRP (DKO) had higher concentrations of FAHFAs particularly isomers with the ester bond at the 9th carbon due to decreased FAHFA hydrolysis activity. The levels of other lipid classes were unaltered indicating that AIG1 and ADTRP specifically hydrolyze FAHFAs. Complementing these genetic studies, we also identified a dual AIG1/ADTRP inhibitor, ABD-110207, which is active in vivo Acute treatment of WT mice with ABD-110207 resulted in elevated FAHFA levels, further supporting the notion that AIG1 and ADTRP activity control endogenous FAHFA levels. However, loss of AIG1/ADTRP did not mimic the changes associated with pharmacologically administered FAHFAs on extent of upregulation of FAHFA levels, glucose tolerance, or insulin sensitivity in mice, indicating that therapeutic strategies should weigh more on FAHFA administration. Together, these findings identify AIG1 and ADTRP as the first endogenous FAHFA hydrolases identified and provide critical genetic and chemical tools for further characterization of these enzymes and endogenous FAHFAs to unravel their physiological functions and roles in health and disease.


Assuntos
Esterases/metabolismo , Ésteres/química , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Proteínas de Membrana/metabolismo , Animais , Esterases/deficiência , Esterases/genética , Técnicas de Inativação de Genes , Hidrólise , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos
7.
Nat Chem Biol ; 16(4): 458-468, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31819274

RESUMO

Functional protein-coding small open reading frames (smORFs) are emerging as an important class of genes. However, the number of translated smORFs in the human genome is unclear because proteogenomic methods are not sensitive enough, and, as we show, Ribo-seq strategies require additional measures to ensure comprehensive and accurate smORF annotation. Here, we integrate de novo transcriptome assembly and Ribo-seq into an improved workflow that overcomes obstacles with previous methods, to more confidently annotate thousands of smORFs. Evolutionary conservation analyses suggest that hundreds of smORF-encoded microproteins are likely functional. Additionally, many smORFs are regulated during fundamental biological processes, such as cell stress. Peptides derived from smORFs are also detectable on human leukocyte antigen complexes, revealing smORFs as a source of antigens. Thus, by including additional validation into our smORF annotation workflow, we accurately identify thousands of unannotated translated smORFs that will provide a rich pool of unexplored, functional human genes.


Assuntos
Anotação de Sequência Molecular/métodos , Fases de Leitura Aberta/genética , Análise de Sequência de DNA/métodos , Genoma Humano , Humanos , Peptídeos/química , Transcriptoma/genética
8.
Nat Commun ; 10(1): 4883, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31653868

RESUMO

Cellular homeostasis relies on having dedicated and coordinated responses to a variety of stresses. The accumulation of unfolded proteins in the endoplasmic reticulum (ER) is a common stress that triggers a conserved pathway called the unfolded protein response (UPR) that mitigates damage, and dysregulation of UPR underlies several debilitating diseases. Here, we discover that a previously uncharacterized 54-amino acid microprotein PIGBOS regulates UPR. PIGBOS localizes to the mitochondrial outer membrane where it interacts with the ER protein CLCC1 at ER-mitochondria contact sites. Functional studies reveal that the loss of PIGBOS leads to heightened UPR and increased cell death. The characterization of PIGBOS reveals an undiscovered role for a mitochondrial protein, in this case a microprotein, in the regulation of UPR originating in the ER. This study demonstrates microproteins to be an unappreciated class of genes that are critical for inter-organelle communication, homeostasis, and cell survival.


Assuntos
Canais de Cloreto/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Proteínas Mitocondriais/metabolismo , Resposta a Proteínas não Dobradas , Animais , Células COS , Morte Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Membranas Mitocondriais/metabolismo , Mapas de Interação de Proteínas , Coelhos , Ratos
9.
Genes Dev ; 33(7-8): 418-435, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30819820

RESUMO

The RNA polymerase II (RNAPII) C-terminal domain kinase, CDK12, regulates genome stability, expression of DNA repair genes, and cancer cell resistance to chemotherapy and immunotherapy. In addition to its role in mRNA biosynthesis of DNA repair genes, we show here that CDK12 phosphorylates the mRNA 5' cap-binding repressor, 4E-BP1, to promote translation of mTORC1-dependent mRNAs. In particular, we found that phosphorylation of 4E-BP1 by mTORC1 (T37 and T46) facilitates subsequent CDK12 phosphorylation at two Ser-Pro sites (S65 and T70) that control the exchange of 4E-BP1 with eIF4G at the 5' cap of CHK1 and other target mRNAs. RNA immunoprecipitation coupled with deep sequencing (RIP-seq) revealed that CDK12 regulates release of 4E-BP1, and binding of eIF4G, to many mTORC1 target mRNAs, including those needed for MYC transformation. Genome-wide ribosome profiling (Ribo-seq) further identified specific CDK12 "translation-only" target mRNAs, including many mTORC1 target mRNAs as well as many subunits of mitotic and centromere/centrosome complexes. Accordingly, confocal imaging analyses revealed severe chromosome misalignment, bridging, and segregation defects in cells deprived of CDK12 or CCNK. We conclude that the nuclear RNAPII-CTD kinase CDK12 cooperates with mTORC1, and controls a specialized translation network that is essential for mitotic chromosome stability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quinase 1 do Ponto de Checagem/genética , Quinases Ciclina-Dependentes/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Instabilidade Genômica/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/genética , Ciclinas/genética , Ciclinas/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Humanos , Mitose/genética , Fosforilação/genética , Ligação Proteica/genética
10.
Biochemistry ; 57(38): 5564-5575, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30215512

RESUMO

Recent technological advances led to the discovery of hundreds to thousands of peptides and small proteins (microproteins) encoded by small open reading frames (smORFs). Characterization of new microproteins demonstrates their role in fundamental biological processes and highlights the value in discovering and characterizing more microproteins. The elucidation of microprotein-protein interactions (MPIs) is useful for determining the biochemical and cellular roles of microproteins. In this study, we characterize the protein interaction partners of mitochondrial elongation factor 1 microprotein (MIEF1-MP) using a proximity labeling strategy that relies on APEX2. MIEF1-MP localizes to the mitochondrial matrix where it interacts with the mitochondrial ribosome (mitoribosome). Functional studies demonstrate that MIEF1-MP regulates mitochondrial translation via its binding to the mitoribosome. Loss of MIEF1-MP decreases the mitochondrial translation rate, while an elevated level of MIEF1-MP increases the translation rate. The identification of MIEF1-MP reveals a new gene involved in this process.


Assuntos
Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Fases de Leitura Aberta , Fatores de Alongamento de Peptídeos/metabolismo , Biossíntese de Proteínas , Sequência de Aminoácidos , Células HEK293 , Células HeLa , Humanos , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Fatores de Alongamento de Peptídeos/genética , Homologia de Sequência
11.
Cell Metab ; 27(2): 419-427.e4, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29414687

RESUMO

Palmitic acid hydroxystearic acids (PAHSAs) are endogenous lipids with anti-diabetic and anti-inflammatory effects. PAHSA levels are reduced in serum and adipose tissue of insulin-resistant people and high-fat diet (HFD)-fed mice. Here, we investigated whether chronic PAHSA treatment enhances insulin sensitivity and which receptors mediate PAHSA effects. Chronic PAHSA administration in chow- and HFD-fed mice raises serum and tissue PAHSA levels ∼1.4- to 3-fold. This improves insulin sensitivity and glucose tolerance without altering body weight. PAHSA administration in chow-fed, but not HFD-fed, mice augments insulin and glucagon-like peptide (GLP-1) secretion. PAHSAs are selective agonists for GPR40, increasing Ca+2 flux, but not intracellular cyclic AMP. Blocking GPR40 reverses improvements in glucose tolerance and insulin sensitivity in PAHSA-treated chow- and HFD-fed mice and directly inhibits PAHSA augmentation of glucose-stimulated insulin secretion in human islets. In contrast, GLP-1 receptor blockade in PAHSA-treated chow-fed mice reduces PAHSA effects on glucose tolerance, but not on insulin sensitivity. Thus, PAHSAs activate GPR40, which is involved in their beneficial metabolic effects.


Assuntos
Glucose/metabolismo , Homeostase , Ácido Palmítico/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Ácidos Esteáricos/farmacologia , Adiposidade/efeitos dos fármacos , Animais , Ingestão de Alimentos/efeitos dos fármacos , Células HEK293 , Homeostase/efeitos dos fármacos , Humanos , Inflamação/patologia , Resistência à Insulina , Camundongos Endogâmicos C57BL
12.
Cell Metab ; 27(1): 218-225.e4, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29103923

RESUMO

Pancreatic α cells retain considerable plasticity and can, under the right circumstances, transdifferentiate into functionally mature ß cells. In search of a targetable mechanistic basis, a recent paper suggested that the widely used anti-malaria drug artemether suppresses the α cell transcription factor Arx to promote transdifferentiation into ß cells. However, key initial experiments in this paper were carried out in islet cell lines, and most subsequent validation experiments implied transdifferentiation without direct demonstration of α to ß cell conversion. Indeed, we find no evidence that artemether promotes transdifferentiation of primary α cells into ß cells. Moreover, artemether reduces Ins2 expression in primary ß cells >100-fold, suppresses glucose uptake, and abrogates ß cell calcium responses and insulin secretion in response to glucose. Our observations suggest that artemether induces general islet endocrine cell dedifferentiation and call into question the utility of artemisinins to promote α to ß cell transdifferentiation in treating diabetes.


Assuntos
Artemeter/farmacologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Transdiferenciação Celular , Células Cultivadas , Células Secretoras de Glucagon/efeitos dos fármacos , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Insulina/metabolismo , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo
13.
Biochemistry ; 56(26): 3299-3306, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28589727

RESUMO

Microproteins are peptides and small proteins encoded by small open reading frames (smORFs). Newer technologies have led to the recent discovery of hundreds to thousands of new microproteins. The biological functions of a few microproteins have been elucidated, and these microproteins have fundamental roles in biology ranging from limb development to muscle function, highlighting the value of characterizing these molecules. The identification of microprotein-protein interactions (MPIs) has proven to be a successful approach to the functional characterization of these genes; however, traditional immunoprecipitation methods result in the enrichment of nonspecific interactions for microproteins. Here, we test and apply an in situ proximity tagging method that relies on an engineered ascorbate peroxidase 2 (APEX) to elucidate MPIs. The results demonstrate that APEX tagging is superior to traditional immunoprecipitation methods for microproteins. Furthermore, the application of APEX tagging to an uncharacterized microprotein called C11orf98 revealed that this microprotein interacts with nucleolar proteins nucleophosmin and nucleolin, demonstrating the ability of this approach to identify novel hypothesis-generating MPIs.


Assuntos
Ascorbato Peroxidases/metabolismo , Nucléolo Celular/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Engenharia de Proteínas , Proteômica/métodos , Proteínas de Ligação a RNA/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Ascorbato Peroxidases/química , Ascorbato Peroxidases/genética , Sequência Conservada , Células HEK293 , Células HeLa , Hemaglutininas/química , Hemaglutininas/genética , Hemaglutininas/metabolismo , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Proteínas Nucleares/química , Proteínas Nucleares/genética , Nucleofosmina , Oligopeptídeos/química , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Nucleolina
14.
Cell Metab ; 25(4): 911-926.e6, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28380380

RESUMO

Postnatal maintenance or regeneration of pancreatic beta cells is considered to occur exclusively via the replication of existing beta cells, but clinically meaningful restoration of human beta cell mass by proliferation has never been achieved. We discovered a population of immature beta cells that is present throughout life and forms from non-beta precursors at a specialized micro-environment or "neogenic niche" at the islet periphery. These cells express insulin, but lack other key beta cell markers, and are transcriptionally immature, incapable of sensing glucose, and unable to support calcium influx. They constitute an intermediate stage in the transdifferentiation of alpha cells to cells that are functionally indistinguishable from conventional beta cells. We thus identified a lifelong source of new beta cells at a specialized site within healthy islets. By comparing co-existing immature and mature beta cells within healthy islets, we stand to learn how to mature insulin-expressing cells into functional beta cells.


Assuntos
Envelhecimento/fisiologia , Microambiente Celular , Células Secretoras de Insulina/citologia , Adulto , Diferenciação Celular/genética , Transdiferenciação Celular , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Perfilação da Expressão Gênica , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Doadores de Tecidos , Transcrição Gênica , Urocortinas/metabolismo
15.
J Biol Chem ; 291(42): 22207-22217, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27573241

RESUMO

We recently discovered a structurally novel class of endogenous lipids, branched palmitic acid esters of hydroxy stearic acids (PAHSAs), with beneficial metabolic and anti-inflammatory effects. We tested whether PAHSAs protect against colitis, which is a chronic inflammatory disease driven predominantly by defects in the innate mucosal barrier and adaptive immune system. There is an unmet clinical need for safe and well tolerated oral therapeutics with direct anti-inflammatory effects. Wild-type mice were pretreated orally with vehicle or 5-PAHSA (10 mg/kg) and 9-PAHSA (5 mg/kg) once daily for 3 days, followed by 10 days of either 0% or 2% dextran sulfate sodium water with continued vehicle or PAHSA treatment. The colon was collected for histopathology, gene expression, and flow cytometry. Intestinal crypt fractions were prepared for ex vivo bactericidal assays. Bone marrow-derived dendritic cells pretreated with vehicle or PAHSA and splenic CD4+ T cells from syngeneic mice were co-cultured to assess antigen presentation and T cell activation in response to LPS. PAHSA treatment prevented weight loss, improved colitis scores (stool consistency, hematochezia, and mouse appearance), and augmented intestinal crypt Paneth cell bactericidal potency via a mechanism that may involve GPR120. In vitro, PAHSAs attenuated dendritic cell activation and subsequent T cell proliferation and Th1 polarization. The anti-inflammatory effects of PAHSAs in vivo resulted in reduced colonic T cell activation and pro-inflammatory cytokine and chemokine expression. These anti-inflammatory effects appear to be partially GPR120-dependent. We conclude that PAHSA treatment regulates innate and adaptive immune responses to prevent mucosal damage and protect against colitis. Thus, PAHSAs may be a novel treatment for colitis and related inflammation-driven diseases.


Assuntos
Imunidade Adaptativa/imunologia , Colite/tratamento farmacológico , Ácidos Graxos/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Celulas de Paneth/imunologia , Células Th1/imunologia , Animais , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Sulfato de Dextrana/farmacologia , Masculino , Camundongos , Celulas de Paneth/patologia , Receptores Acoplados a Proteínas G/imunologia , Células Th1/patologia
16.
Mol Metab ; 5(7): 449-458, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27408771

RESUMO

OBJECTIVE: Complex local crosstalk amongst endocrine cells within the islet ensures tight coordination of their endocrine output. This is illustrated by the recent demonstration that the negative feedback control by delta cells within pancreatic islets determines the homeostatic set-point for plasma glucose during mouse postnatal development. However, the close association of islet endocrine cells that facilitates paracrine crosstalk also complicates the distinction between effects mediated directly on beta cells from indirect effects mediated via local intermediates, such as somatostatin from delta cells. METHODS: To resolve this problem, we generated reporter mice that allow collection of pure pancreatic delta cells along with alpha and beta cells from the same islets and generated comprehensive transcriptomes for each islet endocrine cell type. These transcriptomes afford an unparalleled view of the receptors expressed by delta, alpha and beta cells, and allow the prediction of which signal targets which endocrine cell type with great accuracy. RESULTS: From these transcriptomes, we discovered that the ghrelin receptor is expressed exclusively by delta cells within the islet, which was confirmed by fluorescent in situ hybridization and qPCR. Indeed, ghrelin increases intracellular calcium in delta cells in intact mouse islets, measured by GCaMP6 and robustly potentiates glucose-stimulated somatostatin secretion on mouse and human islets in both static and perfusion assays. In contrast, des-acyl-ghrelin at the same dose had no effect on somatostatin secretion and did not block the actions of ghrelin. CONCLUSIONS: These results offer a straightforward explanation for the well-known insulinostatic actions of ghrelin. Rather than engaging beta cells directly, ghrelin engages delta cells to promote local inhibitory feedback that attenuates insulin release. These findings illustrate the power of our approach to resolve some of the long-standing conundrums with regard to the rich feedback that occurs within the islet that is integral to islet physiology and therefore highly relevant to diabetes.

17.
J Am Chem Soc ; 138(30): 9473-8, 2016 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-27379525

RESUMO

Apolipoprotein E (ApoE) belongs to a large class of proteins that solubilize lipids for physiological transport. Humans have three different APOE alleles, APOE ε2, APOE ε3, and APOE ε4, and genetic studies identified ApoE4 as the strongest genetic risk factor for Alzheimer's disease (AD). People who are homozygous for ApoE4 (i.e., ApoE4/E4) are an order of magnitude more likely to develop late-onset AD (LOAD) than ApoE3/E3 carriers. Several differences between ApoE3 and ApoE4 may contribute to AD including the observation that ApoE4 is degraded to a greater extent than ApoE3 in the human brain. Experiments with high-temperature requirement serine peptidase A1 (HtrA1), which is found in the nervous system, demonstrate that HtrA1 is an allele-selective ApoE-degrading enzyme that degrades ApoE4 more quickly than ApoE3. This activity is specific to HtrA1, as similar assays with HtrA2 showed minimal ApoE4 proteolysis and trypsin had no preference between ApoE4 and ApoE3. HtrA1 has also been reported to cleave the tau protein (Tau) and the amyloid protein precursor (APP) to hinder the formation of toxic amyloid deposits associated with AD. Competition assays with ApoE4, ApoE3, and Tau revealed that ApoE4 inhibits Tau degradation. Thus, the identification of ApoE4 as an in vitro HtrA1 substrate suggests a potential biochemical mechanism that links ApoE4 regulation of AD proteins such as Tau.


Assuntos
Alelos , Apolipoproteínas E/genética , Serina Peptidase 1 de Requerimento de Alta Temperatura A/metabolismo , Proteólise , Células HEK293 , Serina Peptidase 1 de Requerimento de Alta Temperatura A/química , Humanos , Modelos Moleculares , Conformação Proteica
18.
Anal Chem ; 88(7): 3967-75, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27010111

RESUMO

Computational, genomic, and proteomic approaches have been used to discover nonannotated protein-coding small open reading frames (smORFs). Some novel smORFs have crucial biological roles in cells and organisms, which motivates the search for additional smORFs. Proteomic smORF discovery methods are advantageous because they detect smORF-encoded polypeptides (SEPs) to validate smORF translation and SEP stability. Because SEPs are shorter and less abundant than average proteins, SEP detection using proteomics faces unique challenges. Here, we optimize several steps in the SEP discovery workflow to improve SEP isolation and identification. These changes have led to the detection of several new human SEPs (novel human genes), improved confidence in the SEP assignments, and enabled quantification of SEPs under different cellular conditions. These improvements will allow faster detection and characterization of new SEPs and smORFs.


Assuntos
Fases de Leitura Aberta/genética , Peptídeos/análise , Peptídeos/genética , Células HEK293 , Humanos , Células K562 , Peptídeos/isolamento & purificação , Células Tumorais Cultivadas
19.
J Med Chem ; 59(3): 854-66, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26789203

RESUMO

CRF mediates numerous stress-related endocrine, autonomic, metabolic, and behavioral responses. We present the synthesis and chemical and biological properties of astressin B analogues {cyclo(30-33)[D-Phe(12),Nle(21,38),C(α)MeLeu(27,40),Glu(30),Lys(33)]-acetyl-h/r-CRF(9-41)}. Out of 37 novel peptides, 17 (2, 4, 6-8, 10, 11, 16, 17, 27, 29, 30, 32-36) and 16 (3, 5, 9, 12-15, 18, 19, 22-26, 28, 31) had k(i) to CRF receptors in the high picomolar and low nanomole ranges, respectively. Peptides 1, 2, and 11 inhibited h/rCRF and urocortin 1-induced cAMP release from AtT20 and A7r5 cells. When Astressin C 2 was administered to adrenalectomized rats at 1.0 mg subcutaneously, it inhibited ACTH release for >7 d. Additional rat data based on the inhibitory effect of (2) on h/rCRF-induced stimulation of colonic secretory motor activity and urocortin 2-induced delayed gastric emptying also indicate a safe and long-lasting antagonistic effect. The overall properties of selected analogues may fulfill the criteria expected from clinical candidates.


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Animais , Hormônio Liberador da Corticotropina/administração & dosagem , Hormônio Liberador da Corticotropina/química , AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Ratos , Relação Estrutura-Atividade , Urocortinas/antagonistas & inibidores
20.
Nat Med ; 21(7): 769-76, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26076035

RESUMO

The peptide hormone urocortin3 (Ucn3) is abundantly expressed by mature beta cells, yet its physiological role is unknown. Here we demonstrate that Ucn3 is stored and co-released with insulin and potentiates glucose-stimulated somatostatin secretion via cognate receptors on delta cells. Further, we found that islets lacking endogenous Ucn3 have fewer delta cells, reduced somatostatin content, impaired somatostatin secretion, and exaggerated insulin release, and that these defects are rectified by treatment with synthetic Ucn3 in vitro. Our observations indicate that the paracrine actions of Ucn3 activate a negative feedback loop that promotes somatostatin release to ensure the timely reduction of insulin secretion upon normalization of plasma glucose. Moreover, Ucn3 is markedly depleted from beta cells in mouse and macaque models of diabetes and in human diabetic islets. This suggests that Ucn3 is a key contributor to stable glycemic control, whose reduction during diabetes aggravates glycemic volatility and contributes to the pathophysiology of this disease.


Assuntos
Retroalimentação Fisiológica , Insulina/metabolismo , Somatostatina/metabolismo , Urocortinas/metabolismo , Adolescente , Adulto , Idoso , Animais , Criança , Pré-Escolar , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Hiperglicemia/genética , Hiperglicemia/patologia , Lactente , Recém-Nascido , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Macaca , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Modelos Biológicos , Comunicação Parácrina , Doadores de Tecidos , Transcriptoma/genética , Urocortinas/deficiência , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...