Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
medRxiv ; 2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-38014192

RESUMO

Background & Aims: Total proctocolectomy with ileal pouch anal anastomosis (IPAA) is the standard of care for patients with severe treatment resistant ulcerative colitis (UC). Despite improvements in patient outcomes, about 50% of patients will develop inflammation of the pouch within 1-2 years following surgery. Establishment of UC pouches is associated with profound histological changes of the mucosa. A detailed characterization of these changes on a cellular and molecular level is crucial for an improved understanding of pouch physiology and diseases management. Methods: We generated cell-type-resolved transcriptional and epigenetic atlases of UC pouches using scRNA-seq and scATAC-seq data from paired biopsy samples from the ileal pouch and ileal segment above the pouch (pre-pouch) of UC-IPAA patients (n=6, female=2) without symptoms. We also collected data from paired biopsies of the terminal ileum (TI) and ascending colon (AC) from healthy controls (n=6, female=3). Results: We identified novel populations of colon-like absorptive and secretory epithelial cells, constituting a significant proportion of the epithelial cell fraction in the pouch but not in matched pre-pouch samples. Pouch-specific enterocytes expressed colon-specific genes, including CEACAM5, CA2. However, in contrast to normal colonic epithelium, these cells also expressed a range of inflammatory and secretory genes, similar to previously detected gene expression signatures in IBD patients. Comparison to longitudinal bulk RNA-seq data from UC pouches demonstrated that colon-like epithelial cells are present early after pouch functionalization and independently of subsequent pouchitis. Finally, single cell chromatin accessibility revealed activation colonic transcriptional regulators, including CDX1, NFIA, and EHF. Conclusion: UC pouches are characterized by partial colonic metaplasia of the epithelium. These data constitute a resource of transcriptomic and epigenetic signatures of cell populations in the pouch and provide an anchor for understanding the underlying molecular mechanisms of pouchitis.

2.
Science ; 381(6657): 502-508, 2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37535745

RESUMO

The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.


Assuntos
Antifúngicos , Peptídeos Antimicrobianos , Candida , Celulas de Paneth , Fragmentos de Peptídeos , Peptídeo YY , Animais , Antifúngicos/metabolismo , Peptídeos Antimicrobianos/metabolismo , Candida/efeitos dos fármacos , Candida/fisiologia , Celulas de Paneth/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeo YY/metabolismo , Simbiose , Humanos , Camundongos
3.
Cell Host Microbe ; 30(6): 809-823.e6, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35439436

RESUMO

Gut microbial diurnal oscillations are important diet-dependent drivers of host circadian rhythms and metabolism ensuring optimal energy balance. However, the interplay between diet, microbes, and host factors sustaining intestinal oscillations is complex and poorly understood. Here, using a mouse model, we report the host C-type lectin antimicrobial peptide Reg3γ works with key ileal microbes to orchestrate these interactions in a bidirectional manner and does not correlate with the intestinal core circadian clock. High-fat diet is the primary driver of microbial oscillators that impair host metabolic homeostasis, resulting in arrhythmic host Reg3γ expression that secondarily drives abundance and oscillation of key gut microbes. This illustrates transkingdom coordination of biological rhythms primarily influenced by diet and reciprocal sensor-effector signals between host and microbial components, ultimately driving metabolism. Restoring the gut microbiota's capacity to sense dietary signals mediated by specific host factors such as Reg3γ could be harnessed to improve metabolic dysfunction.


Assuntos
Relógios Circadianos , Microbioma Gastrointestinal , Ritmo Circadiano , Dieta , Dieta Hiperlipídica/efeitos adversos , Metabolismo dos Lipídeos
4.
Transl Res ; 236: 35-51, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34126257

RESUMO

Patients with one of the many chronic inflammatory disorders broadly classified as inflammatory bowel disease (IBD) now have a diverse set of immunomodulatory therapies at their disposal. Despite these recent medical advances, complete sustained remission of disease remains elusive for most patients. The full healing of the damaged intestinal mucosa is the primary goal of all therapies. Achieving this requires not just a reduction of the aberrant immunological response, but also wound healing of the epithelium. No currently approved therapy directly targets the epithelium. Epithelial repair is compromised in IBD and normally facilitates re-establishment of the homeostatic barrier between the host and the microbiome. In this review, we summarize the evidence that epithelial wound healing represents an important yet underdeveloped therapeutic modality for IBD. We highlight 3 general approaches that are promising for developing a new class of epithelium-targeted therapies: epithelial stem cells, cytokines, and microbiome engineering. We also provide a frank discussion of some of the challenges that must be overcome for epithelial repair to be therapeutically leveraged. A concerted approach by the field to develop new therapies targeting epithelial wound healing will offer patients a game-changing, complementary class of medications and could dramatically improve outcomes.


Assuntos
Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/terapia , Mucosa Intestinal/patologia , Cicatrização , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/microbiologia , Regeneração , Células-Tronco/patologia
5.
Elife ; 92020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32553109

RESUMO

Traditional cultivation approaches in microbiology are labor-intensive, low-throughput, and yield biased sampling of environmental microbes due to ecological and evolutionary factors. New strategies are needed for ample representation of rare taxa and slow-growers that are often outcompeted by fast-growers in cultivation experiments. Here we describe a microfluidic platform that anaerobically isolates and cultivates microbial cells in millions of picoliter droplets and automatically sorts them based on colony density to enhance slow-growing organisms. We applied our strategy to a fecal microbiota transplant (FMT) donor stool using multiple growth media, and found significant increase in taxonomic richness and larger representation of rare and clinically relevant taxa among droplet-grown cells compared to conventional plates. Furthermore, screening the FMT donor stool for antibiotic resistance revealed 21 populations that evaded detection in plate-based assessment of antibiotic resistance. Our method improves cultivation-based surveys of diverse microbiomes to gain deeper insights into microbial functioning and lifestyles.


The human gut is inhabited with hundreds of billions of bacterial cells from a wide range of families. This complex mixture of bacteria is part of the gut microbiome, along with other lifeforms such as viruses, archaea and fungi. As well as interacting with each other, the bacteria in the microbiome interact with our cells and available nutrients. Studying these interactions can help us understand how this community of bacteria influence health and disease. One way to study the diversity of the microbiome is to take a sample, such as a section of stool, and perform DNA sequencing to determine which types of bacteria are present. This can reveal how the composition of the gut microbiome relates to our health, but cannot confirm whether these bacteria are the cause or the effect of most diseases. To overcome this problem, researchers need to be able to grow pure strains of these bacteria in order to unravel their underlying mechanisms. For over a century, the conventional way to cultivate bacteria has been to grow them in a Petri dish. However, this method promotes the growth of more abundant, fast-growing bacterial strains. This results in a huge disconnect between the bacteria grown in a Petri dish and the diversity within the human gut, which is hindering our understanding of gut health and disease. Now, Watterson et al. have built a machine that improves the speed and number of cultivated bacterial organisms, thus paving the way for more detailed investigations of the human gut microbiome. This new system works by growing bacteria in millions of miniscule droplets which can be physically separated to help the expansion of slower growing species. Watterson et al. cultivated bacterial cells from a stool sample from a single donor using the droplet system and compared this to traditional culturing methods. The droplet technology increased the number of different organisms that were able to grow by up to four times, including those that were rare or slow-growing. Bacteria in the donor stool were then screened for populations that were resistant to antibiotics. This identified 21 antibiotic resistant bacteria which only grew in the droplets and not in Petri dishes. This droplet-based technology will make it possible to study bacterial strains that were previously difficult to grow. Furthermore, this method could help identify whether stool from a donor contains any antibiotic resistant strains, which can lead to clinical complications once transplanted. In future, this new technology could be used in laboratories or hospitals to study the role of the gut microbiome in health and disease.


Assuntos
Bactérias/efeitos dos fármacos , Técnicas Bacteriológicas/métodos , Farmacorresistência Bacteriana , Microbioma Gastrointestinal , Ensaios de Triagem em Larga Escala/métodos , Técnicas Bacteriológicas/instrumentação , Ensaios de Triagem em Larga Escala/instrumentação
6.
Cell Rep ; 25(7): 1816-1828.e4, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30428350

RESUMO

Transcriptional regulation of circadian rhythms is essential for lipid metabolic homeostasis, disruptions of which can lead to metabolic diseases. Whether N6-methyladenosine (m6A) mRNA methylation impacts circadian regulation of lipid metabolism is unclear. Here, we show m6A mRNA methylation oscillations in murine liver depend upon a functional circadian clock. Hepatic deletion of Bmal1 increases m6A mRNA methylation, particularly of PPaRα. Inhibition of m6A methylation via knockdown of m6A methyltransferase METTL3 decreases PPaRα m6A abundance and increases PPaRα mRNA lifetime and expression, reducing lipid accumulation in cells in vitro. Mechanistically, YTHDF2 binds to PPaRα to mediate its mRNA stability to regulate lipid metabolism. Induction of reactive oxygen species both in vitro and in vivo increases PPaRα transcript m6A levels, revealing a possible mechanism for circadian disruption on m6A mRNA methylation. These data show that m6A RNA methylation is important for circadian regulation of downstream genes and lipid metabolism, impacting metabolic outcomes.


Assuntos
Adenosina/análogos & derivados , Relógios Circadianos/genética , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Fatores de Transcrição ARNTL/metabolismo , Adenosina/metabolismo , Animais , Proliferação de Células , Deleção de Genes , Células Hep G2 , Humanos , Metilação , Metiltransferases/metabolismo , Camundongos Knockout , Modelos Biológicos , PPAR alfa/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
7.
Cell Host Microbe ; 23(4): 458-469.e5, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29649441

RESUMO

The gut microbiota play important roles in lipid metabolism and absorption. However, the contribution of the small bowel microbiota of mammals to these diet-microbe interactions remains unclear. We determine that germ-free (GF) mice are resistant to diet-induced obesity and malabsorb fat with specifically impaired lipid digestion and absorption within the small intestine. Small bowel microbes are essential for host adaptation to dietary lipid changes by regulating gut epithelial processes involved in their digestion and absorption. In addition, GF mice conventionalized with high-fat diet-induced jejunal microbiota exhibit increased lipid absorption even when fed a low-fat diet. Conditioned media from specific bacterial strains directly upregulate lipid absorption genes in murine proximal small intestinal epithelial organoids. These findings indicate that proximal gut microbiota play key roles in host adaptability to dietary lipid variations through mechanisms involving both the digestive and absorptive phases and that these functions may contribute to conditions of over- and undernutrition.


Assuntos
Dieta/métodos , Microbioma Gastrointestinal , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Metabolismo dos Lipídeos , Animais , Camundongos
9.
Chem Sci ; 8(11): 7588-7592, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29568422

RESUMO

The reversible modification of cysteine residues through thioester formation with palmitate (protein S-palmitoylation) is a prevalent chemical modification that regulates the function, localization, and stability of many proteins. Current methods for monitoring the "erasers" of S-palmitoylation, acyl-protein thioesterases (APTs), rely on destructive proteomic methods or "turn-on" probes, precluding deployment in heterogeneous samples such as primary tissues. To address these challenges, we present the design, synthesis, and biological evaluation of Ratiometric Depalmitoylation Probes (RDPs). RDPs respond to APTs with a robust ratiometric change in fluorescent signal both in vitro and in live cells. Moreover, RDPs can monitor endogenous APT activities in heterogeneous primary human tissues such as colon organoids, presaging the utility of these molecules in uncovering novel roles for APTs in metabolic regulation.

10.
Am J Physiol Gastrointest Liver Physiol ; 309(6): G431-42, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26185331

RESUMO

Stimulation of digestive organs by enteric peptides is lost during total parental nutrition (PN). Here we examine the role of the enteric peptide bombesin (BBS) in stimulation of the exocrine and endocrine pancreas during PN. BBS protects against exocrine pancreas atrophy and dysfunction caused by PN. BBS also augments circulating insulin levels, suggesting an endocrine pancreas phenotype. While no significant changes in gross endocrine pancreas morphology were observed, pancreatic islets isolated from BBS-treated PN mice showed a significantly enhanced insulin secretion response to the glucagon-like peptide-1 (GLP-1) agonist exendin-4, correlating with enhanced GLP-1 receptor expression. BBS itself had no effect on islet function, as reflected in low expression of BBS receptors in islet samples. Intestinal BBS receptor expression was enhanced in PN with BBS, and circulating active GLP-1 levels were significantly enhanced in BBS-treated PN mice. We hypothesized that BBS preserved islet function indirectly, through the enteroendocrine cell-pancreas axis. We confirmed the ability of BBS to directly stimulate intestinal enteroid cells to express the GLP-1 precursor preproglucagon. In conclusion, BBS preserves the exocrine and endocrine pancreas functions during PN; however, the endocrine stimulation is likely indirect, through the enteroendocrine cell-pancreas axis.


Assuntos
Bombesina/farmacologia , Peptídeo Liberador de Gastrina/análogos & derivados , Ilhotas Pancreáticas/efeitos dos fármacos , Pâncreas Exócrino/efeitos dos fármacos , Nutrição Parenteral/efeitos adversos , Amilases/metabolismo , Animais , DNA/metabolismo , Alimentos Formulados , Regulação da Expressão Gênica , Hiperglicemia/sangue , Ilhotas Pancreáticas/anatomia & histologia , Lipase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Pâncreas Exócrino/anatomia & histologia , Hormônios Pancreáticos/metabolismo
11.
Cell Host Microbe ; 17(5): 681-9, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25891358

RESUMO

Circadian clocks and metabolism are inextricably intertwined, where central and hepatic circadian clocks coordinate metabolic events in response to light-dark and sleep-wake cycles. We reveal an additional key element involved in maintaining host circadian rhythms, the gut microbiome. Despite persistence of light-dark signals, germ-free mice fed low or high-fat diets exhibit markedly impaired central and hepatic circadian clock gene expression and do not gain weight compared to conventionally raised counterparts. Examination of gut microbiota in conventionally raised mice showed differential diurnal variation in microbial structure and function dependent upon dietary composition. Additionally, specific microbial metabolites induced under low- or high-fat feeding, particularly short-chain fatty acids, but not hydrogen sulfide, directly modulate circadian clock gene expression within hepatocytes. These results underscore the ability of microbially derived metabolites to regulate or modify central and hepatic circadian rhythm and host metabolic function, the latter following intake of a Westernized diet.


Assuntos
Relógios Circadianos , Dieta Hiperlipídica , Disbiose/induzido quimicamente , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Metabolismo dos Lipídeos , Animais , Peso Corporal , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Fígado/patologia , Camundongos , Dados de Sequência Molecular , Obesidade , Análise de Sequência de DNA
12.
J Clin Invest ; 125(3): 1098-110, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25642769

RESUMO

The intracellular protein HMGB1 is released from cells and acts as a damage-associated molecular pattern molecule during many diseases, including inflammatory bowel disease (IBD); however, the intracellular function of HMGB1 during inflammation is poorly understood. Here, we demonstrated that cytosolic HMGB1 regulates apoptosis by protecting the autophagy proteins beclin 1 and ATG5 from calpain-mediated cleavage during inflammation. Colitis in mice with an intestinal epithelial cell-specific Hmgb1 deletion and patients with IBD were both characterized by increased calpain activation, beclin 1 and ATG5 cleavage, and intestinal epithelial cell (IEC) death compared with controls. In vitro cleavage assays and studies of enteroids verified that HMGB1 protects beclin 1 and ATG5 from calpain-mediated cleavage events that generate proapoptotic protein fragments. Together, our results indicate that HMGB1 is essential for mitigating the extent and severity of inflammation-associated cellular injury by controlling the switch between the proautophagic and proapoptotic functions of beclin 1 and ATG5 during inflammation. Moreover, these studies demonstrate that HMGB1 is pivotal for reducing tissue injury in IBD and other complex inflammatory disorders.


Assuntos
Apoptose , Autofagia , Colite/metabolismo , Proteína HMGB1/fisiologia , Imunidade Adaptativa , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 5 Relacionada à Autofagia , Proteína Beclina-1 , Calpaína/antagonistas & inibidores , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Citosol/metabolismo , Sulfato de Dextrana , Dipeptídeos/farmacologia , Células Epiteliais/fisiologia , Feminino , Humanos , Mucosa Intestinal/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteólise
13.
Curr Opin Immunol ; 31: 16-23, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25214301

RESUMO

Autoimmune disorders, particularly inflammatory bowel diseases (IBD), are increasing at an alarming frequency. While the exact cause remains elusive, studies have examined how the immune system is shaped in the context of genetic susceptibility, gut microbes, and environmental pressures, including dietary intake. Shifts towards a Westernized high fat, high carbohydrate diet result in changes to gut microbiota structure and function that may aid in triggering and perpetuating autoimmunity by promoting the emergence of pathobionts leading to altered immune activation. This review summarizes our current understanding of dietary-induced changes in gut microbiota on autoimmunity in the context of IBD. We provide a framework for leveraging this knowledge to develop new dietary, microbial and immune-based modulation strategies for individualized risk assessment and improving clinical outcomes.


Assuntos
Autoimunidade , Dieta , Doenças Inflamatórias Intestinais , Intestinos , Microbiota/imunologia , Animais , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia
14.
Clin Dev Immunol ; 2012: 780436, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251221

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multiple genetic risk factors, high levels of interferon alpha (IFN-α), and the production of autoantibodies against components of the cell nucleus. Interferon regulatory factor 5 (IRF5) is a transcription factor which induces the transcription of IFN-α and other cytokines, and genetic variants of IRF5 have been strongly linked to SLE pathogenesis. IRF5 functions downstream of Toll-like receptors and other microbial pattern-recognition receptors, and immune complexes made up of SLE-associated autoantibodies seem to function as a chronic endogenous stimulus to this pathway. In this paper, we discuss the physiologic role of IRF5 in immune defense and the ways in which IRF5 variants may contribute to the pathogenesis of human SLE. Recent data regarding the role of IRF5 in both serologic autoimmunity and the overproduction of IFN-α in human SLE are summarized. These data support a model in which SLE-risk variants of IRF5 participate in a "feed-forward" mechanism, predisposing to SLE-associated autoantibody formation, and subsequently facilitating IFN-α production downstream of Toll-like receptors stimulated by immune complexes composed of these autoantibodies.


Assuntos
Doenças Autoimunes/imunologia , Fatores Reguladores de Interferon/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Autoanticorpos/imunologia , Autoimunidade/imunologia , Humanos
15.
Eur J Immunol ; 38(9): 2438-50, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18792400

RESUMO

We recently reported that differentiation of CD8(+) T cells from the naïve to the effector state involves the upregulation of glucose-dependent metabolism. Glucose deprivation or inhibition of glycolysis by 2-deoxy-D-glucose (2-DG) selectively inhibited production of IFN-gamma but not of IL-2. To determine a more global role of glucose metabolism on effector T-cell function, we performed gene array analysis on CD8(+) effector T cells stimulated in the presence or absence of 2-DG. We observed that expression of only 10% of genes induced by TCR/CD28 signaling was inhibited by 2-DG. Among these were genes for key cytokines, cell cycle molecules, and cytotoxic granule proteins. Consistent with these results, production of IFN-gamma and GM-CSF, cell cycle progression, upregulation of cyclin D2 protein, cytolytic activity, and upregulation of granzyme B protein and also conjugate formation were exquisitely glucose-dependent. In contrast to glucose, oxygen was little utilized by CD8(+) effector T cells, and relative oxygen deprivation did not inhibit these CTL functional properties. Our results indicate a particularly critical role for glucose in regulating specific effector functions of CD8(+) T cells and have implications for the maintenance of the effector phase of cellular immune responses in target tissue microenvironments such as a solid tumor.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/biossíntese , Citotoxicidade Imunológica , Expressão Gênica , Glucose/metabolismo , Ativação Linfocitária , Animais , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células , Citocinas/imunologia , Desoxiglucose/farmacologia , Perfilação da Expressão Gênica , Camundongos , Oxigênio/metabolismo
16.
J Immunol ; 174(8): 4670-7, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15814691

RESUMO

Differentiation of CD8+ T cells from the naive to the effector state is accompanied by changes in basal gene expression profiles that parallel the acquisition of effector functions. Among these are metabolism genes, and we now show that 2C TCR transgenic effector CD8+ T cells express higher levels of glycolytic enzymes and display greater glucose uptake, a higher glycolytic rate, and increased lactate production compared with naive cells. To determine whether glucose was required for effector T cell functions, we regulated glucose availability in vitro. Glucose deprivation strongly inhibited IFN-gamma gene expression, whereas IL-2 production was little affected. Inhibition correlated with diminished phosphorylation of p70S6 kinase and eIF4E binding protein 1 and a requirement for de novo protein synthesis, whereas other signaling pathways known to regulate IFN-gamma expression were unaffected. Together, our data reveal that optimal induction of IFN-gamma transcription is a glucose-dependent process, indicate that there are undefined factors that influence IFN-gamma expression, and have implications for regulation of the effector phase of CD8+ T cell responses in tissue microenvironments.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Glucose/metabolismo , Interferon gama/biossíntese , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , DNA/genética , Desoxiglucose/farmacologia , Ativação Enzimática , Fatores de Iniciação em Eucariotos , Regulação da Expressão Gênica , Glicólise/genética , Técnicas In Vitro , Interferon gama/genética , Interleucina-2/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Fosfoproteínas/metabolismo , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Regulação para Cima
17.
Immunol Res ; 31(2): 107-18, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15778509

RESUMO

Established tumors develop ways to elude destruction by the host immune system. Recent work has revealed that tumors can take advantage of the generation of metabolic dysregulation to inhibit immune responses. Effector T-cell functions are particularly sensitive to nutrient availability in the tumor microenvironment. In this review, we highlight experimental data supporting the importance of glucose, oxygen, tryptophan, and arginine for optimal T-cell function, and the mechanisms by which these nutrients may become depleted in the tumor microenvironment. These observations provide a conceptual framework for modulating metabolic features of the T cell-tumor interaction, toward the end of promoting more effective immune-mediated tumor destruction in vivo.


Assuntos
Movimento Celular/imunologia , Tolerância Imunológica , Neoplasias/imunologia , Neoplasias/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Aminoácidos/deficiência , Aminoácidos/metabolismo , Aminoácidos/fisiologia , Animais , Humanos , Neoplasias/patologia , Neoplasias/prevenção & controle , Subpopulações de Linfócitos T/citologia
18.
J Biol Chem ; 278(19): 17044-52, 2003 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-12582156

RESUMO

Peripheral CD8(+) T cells circulate in a quiescent naive state until they are primed by specific antigen and differentiate into effector cells. In the effector state, CD8(+) T cells acquire cytolytic activity and produce increased levels of cytokines such as interferon-gamma. They also exhibit increased T cell receptor sensitivity, decreased CD28 dependence, and become inhibitable by CTLA-4 and other negative regulatory pathways. We hypothesized that one mechanism by which these two states are regulated is via differential expression of specific genes. To this end, basal gene expression profiles of naive and effector 2C TCR transgenic x RAG2(-/-) CD8(+) T cells were analyzed using Affymetrix arrays representing 11,000 genes. Of the 177 differentially expressed known genes, 68 were expressed at higher levels in effector cells, but 109 were more abundant in naive cells, supporting the notion that the naive state is not passive. Expression of genes related to metabolism, actin cytoskeletal dynamics, and effector function increased with priming, whereas expression of putative anti-proliferative genes decreased. Semiquantitative reverse transcription-PCR was utilized as a secondary validation for selected transcripts, and Western blot analysis was used to examine protein expression for molecules of interest. Surprisingly, for 24 genes examined, 12 showed discordant protein versus mRNA expression. In summary, our study indicates that: 1) not only does the expression of some genes in naive CD8(+) T cells become up-regulated upon priming, but the expression of other genes is down-regulated as well and 2) the complexities of T cell differentiation include regulation at the post-transcriptional level.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Diferenciação Celular/fisiologia , Citocinas/biossíntese , Processamento Pós-Transcricional do RNA , Animais , Antígenos CD8 , Citocinas/genética , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , RNA/análise , RNA/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...