Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 157(7): 1605-18, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24949972

RESUMEN

Translational readthrough, observed primarily in less complex organisms from viruses to Drosophila, expands the proteome by translating select transcripts beyond the canonical stop codon. Here, we show that vascular endothelial growth factor A (VEGFA) mRNA in mammalian endothelial cells undergoes programmed translational readthrough (PTR) generating VEGF-Ax, an isoform containing a unique 22-amino-acid C terminus extension. A cis-acting element in the VEGFA 3' UTR serves a dual function, not only encoding the appended peptide but also directing the PTR by decoding the UGA stop codon as serine. Heterogeneous nuclear ribonucleoprotein (hnRNP) A2/B1 binds this element and promotes readthrough. Remarkably, VEGF-Ax exhibits antiangiogenic activity in contrast to the proangiogenic activity of VEGF-A. Pathophysiological significance of VEGF-Ax is indicated by robust expression in multiple human tissues but depletion in colon adenocarcinoma. Furthermore, genome-wide analysis revealed AGO1 and MTCH2 as authentic readthrough targets. Overall, our studies reveal a novel protein-regulated PTR event in a vertebrate system.


Asunto(s)
Células Endoteliales/metabolismo , Biosíntesis de Proteínas , Factor A de Crecimiento Endotelial Vascular/genética , Regiones no Traducidas 3' , Secuencia de Aminoácidos , Animales , Aorta/citología , Secuencia de Bases , Bovinos , Línea Celular , Codón de Terminación , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Alineación de Secuencia
2.
Cell ; 149(1): 88-100, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22386318

RESUMEN

Posttranscriptional regulatory mechanisms superimpose "fine-tuning" control upon "on-off" switches characteristic of gene transcription. We have exploited computational modeling with experimental validation to resolve an anomalous relationship between mRNA expression and protein synthesis. The GAIT (gamma-interferon-activated inhibitor of translation) complex repressed VEGF-A synthesis to a low, constant rate independent of VEGF-A mRNA expression levels. Dynamic model simulations predicted an inhibitory GAIT-element-interacting factor to account for this relationship and led to the identification of a truncated form of glutamyl-prolyl tRNA synthetase (EPRS), a GAIT constituent that mediates binding to target transcripts. The truncated protein, EPRS(N1), shields GAIT-element-bearing transcripts from the inhibitory GAIT complex, thereby dictating a "translational trickle" of GAIT target proteins. EPRS(N1) mRNA is generated by polyadenylation-directed conversion of a Tyr codon in the EPRS-coding sequence to a stop codon (PAY(∗)). Genome-wide analysis revealed multiple candidate PAY(∗) targets, including the authenticated target RRM1, suggesting a general mechanism for production of C terminus-truncated regulatory proteins.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Regulación de la Expresión Génica , Genoma Humano , Biosíntesis de Proteínas , Secuencia de Aminoácidos , Aminoacil-ARNt Sintetasas/química , Codón de Terminación , Humanos , Leucocitos Mononucleares/metabolismo , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Poliadenilación , Transcriptoma , Células U937 , Factor A de Crecimiento Endotelial Vascular/genética
3.
Gut ; 72(11): 2068-2080, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37080587

RESUMEN

OBJECTIVE: Perianal Crohn's disease (pCD) occurs in up to 40% of patients with CD and is associated with poor quality of life, limited treatment responses and poorly understood aetiology. We performed a genetic association study comparing CD subjects with and without perianal disease and subsequently performed functional follow-up studies for a pCD associated SNP in Complement Factor B (CFB). DESIGN: Immunochip-based meta-analysis on 4056 pCD and 11 088 patients with CD from three independent cohorts was performed. Serological and clinical variables were analysed by regression analyses. Risk allele of rs4151651 was introduced into human CFB plasmid by site-directed mutagenesis. Binding of recombinant G252 or S252 CFB to C3b and its cleavage was determined in cell-free assays. Macrophage phagocytosis in presence of recombinant CFB or serum from CFB risk, or protective CD or healthy subjects was assessed by flow cytometry. RESULTS: Perianal complications were associated with colonic involvement, OmpC and ASCA serology, and serology quartile sum score. We identified a genetic association for pCD (rs4151651), a non-synonymous SNP (G252S) in CFB, in all three cohorts. Recombinant S252 CFB had reduced binding to C3b, its cleavage was impaired, and complement-driven phagocytosis and cytokine secretion were reduced compared with G252 CFB. Serine 252 generates a de novo glycosylation site in CFB. Serum from homozygous risk patients displayed significantly decreased macrophage phagocytosis compared with non-risk serum. CONCLUSION: pCD-associated rs4151651 in CFB is a loss-of-function mutation that impairs its cleavage, activation of alternative complement pathway, and pathogen phagocytosis thus implicating the alternative complement pathway and CFB in pCD aetiology.


Asunto(s)
Factor B del Complemento , Enfermedad de Crohn , Humanos , Factor B del Complemento/genética , Enfermedad de Crohn/complicaciones , Calidad de Vida , Estudios de Seguimiento , Fagocitosis
4.
Nature ; 542(7641): 357-361, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28178239

RESUMEN

Metabolic pathways that contribute to adiposity and ageing are activated by the mammalian target of rapamycin complex 1 (mTORC1) and p70 ribosomal protein S6 kinase 1 (S6K1) axis. However, known mTORC1-S6K1 targets do not account for observed loss-of-function phenotypes, suggesting that there are additional downstream effectors of this pathway. Here we identify glutamyl-prolyl-tRNA synthetase (EPRS) as an mTORC1-S6K1 target that contributes to adiposity and ageing. Phosphorylation of EPRS at Ser999 by mTORC1-S6K1 induces its release from the aminoacyl tRNA multisynthetase complex, which is required for execution of noncanonical functions of EPRS beyond protein synthesis. To investigate the physiological function of EPRS phosphorylation, we generated Eprs knock-in mice bearing phospho-deficient Ser999-to-Ala (S999A) and phospho-mimetic (S999D) mutations. Homozygous S999A mice exhibited low body weight, reduced adipose tissue mass, and increased lifespan, similar to S6K1-deficient mice and mice with adipocyte-specific deficiency of raptor, an mTORC1 constituent. Substitution of the EprsS999D allele in S6K1-deficient mice normalized body mass and adiposity, indicating that EPRS phosphorylation mediates S6K1-dependent metabolic responses. In adipocytes, insulin stimulated S6K1-dependent EPRS phosphorylation and release from the multisynthetase complex. Interaction screening revealed that phospho-EPRS binds SLC27A1 (that is, fatty acid transport protein 1, FATP1), inducing its translocation to the plasma membrane and long-chain fatty acid uptake. Thus, EPRS and FATP1 are terminal mTORC1-S6K1 axis effectors that are critical for metabolic phenotypes.


Asunto(s)
Adiposidad , Aminoacil-ARNt Sintetasas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adipocitos/metabolismo , Envejecimiento/metabolismo , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/genética , Animales , Peso Corporal , Membrana Celular/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Femenino , Insulina/metabolismo , Longevidad/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Mutación , Tamaño de los Órganos , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Transporte de Proteínas , Proteína Reguladora Asociada a mTOR , Proteínas Quinasas S6 Ribosómicas 90-kDa/deficiencia
5.
Gastroenterology ; 160(3): 809-822.e7, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33160965

RESUMEN

BACKGROUND AND AIMS: The host receptor for severe acute respiratory syndrome coronavirus 2, angiotensin-converting enzyme 2 (ACE2), is highly expressed in small bowel (SB). Our aim was to identify factors influencing intestinal ACE2 expression in Crohn's disease (CD), ulcerative colitis (UC), and non-inflammatory bowel disease (IBD) controls. METHODS: Using bulk RNA sequencing or microarray transcriptomics from tissue samples (4 SB and 2 colonic cohorts; n = 495; n = 387 UC; n = 94 non-IBD), we analyzed the relationship between ACE2 with demographics and disease activity and prognosis. We examined the outcome of anti-tumor necrosis factor and anti-interleukin-12/interleukin-23 treatment on SB and colonic ACE2 expression in 3 clinical trials. Univariate and multivariate regression models were fitted. RESULTS: ACE2 levels were consistently reduced in SB CD and elevated in colonic UC compared with non-IBD controls. Elevated SB ACE2 was also associated with demographic features (age and elevated body mass index) associated with poor coronavirus disease 2019 outcomes. Within CD, SB ACE2 was reduced in patients subsequently developing complicated disease. Within UC, colonic ACE2 was elevated in active disease and in patients subsequently requiring anti-tumor necrosis factor rescue therapy. SB and colonic ACE2 expression in active CD and UC were restored by anti-cytokine therapy, most notably in responders. CONCLUSIONS: Reduced SB but elevated colonic ACE2 levels in IBD are associated with inflammation and severe disease, but normalized after anti-cytokine therapy, suggesting compartmentalization of ACE2-related biology in SB and colonic inflammation. The restoration of ACE2 expression with anti-cytokine therapy might be important in the context of severe acute respiratory syndrome coronavirus 2 infection and potentially explain reports of reduced morbidity from coronavirus disease 2019 in IBD patients treated with anti-cytokines.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antiinflamatorios/uso terapéutico , Colitis Ulcerosa/tratamiento farmacológico , Enfermedad de Crohn/tratamiento farmacológico , Intestinos/efectos de los fármacos , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/genética , Antiinflamatorios/efectos adversos , COVID-19/enzimología , COVID-19/inmunología , COVID-19/virología , Estudios de Casos y Controles , Niño , Preescolar , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/enzimología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Bases de Datos Genéticas , Femenino , Regulación Enzimológica de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Intestinos/enzimología , Intestinos/inmunología , Masculino , Persona de Mediana Edad , América del Norte , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Virales/metabolismo , SARS-CoV-2/enzimología , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Inhibidores del Factor de Necrosis Tumoral/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
6.
J Biol Chem ; 293(49): 19148-19156, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30309984

RESUMEN

About 1 billion years ago, in a single-celled holozoan ancestor of all animals, a gene fusion of two tRNA synthetases formed the bifunctional enzyme, glutamyl-prolyl-tRNA synthetase (EPRS). We propose here that a confluence of metabolic, biochemical, and environmental factors contributed to the specific fusion of glutamyl- (ERS) and prolyl- (PRS) tRNA synthetases. To test this idea, we developed a mathematical model that centers on the precursor-product relationship of glutamic acid and proline, as well as metabolic constraints on free glutamic acid availability near the time of the fusion event. Our findings indicate that proline content increased in the proteome during the emergence of animals, thereby increasing demand for free proline. Together, these constraints contributed to a marked cellular depletion of glutamic acid and its products, with potentially catastrophic consequences. In response, an ancient organism invented an elegant solution in which genes encoding ERS and PRS fused to form EPRS, forcing coexpression of the two enzymes and preventing lethal dysregulation. The substantial evolutionary advantage of this coregulatory mechanism is evidenced by the persistence of EPRS in nearly all extant animals.


Asunto(s)
Aminoacil-ARNt Sintetasas/química , Proteínas Bacterianas/química , Evolución Molecular , Modelos Químicos , Aminoacil-ARNt Sintetasas/genética , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Bacterias/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ciclo del Ácido Cítrico , Fusión Génica , Glutamato-ARNt Ligasa/química , Glutamato-ARNt Ligasa/genética , Glutamato-ARNt Ligasa/metabolismo , Ácido Glutámico/química , Ácido Glutámico/metabolismo , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Prolina/química , Prolina/metabolismo , Biosíntesis de Proteínas/genética
7.
PLoS Comput Biol ; 14(4): e1006060, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29659573

RESUMEN

Iron plays vital roles in the human body including enzymatic processes, oxygen-transport via hemoglobin and immune response. Iron metabolism is characterized by ~95% recycling and minor replenishment through diet. Anemia of chronic kidney disease (CKD) is characterized by a lack of synthesis of erythropoietin leading to reduced red blood cell (RBC) formation and aberrant iron recycling. Treatment of CKD anemia aims to normalize RBC count and serum hemoglobin. Clinically, the various fluxes of iron transport and accumulation are not measured so that changes during disease (e.g., CKD) and treatment are unknown. Unwanted iron accumulation in patients is known to lead to adverse effects. Current whole-body models lack the mechanistic details of iron transport related to RBC maturation, transferrin (Tf and TfR) dynamics and assume passive iron efflux from macrophages. Hence, they are not predictive of whole-body iron dynamics and cannot be used to design individualized patient treatment. For prediction, we developed a mechanistic, multi-scale computational model of whole-body iron metabolism incorporating four compartments containing major pools of iron and RBC generation process. The model accounts for multiple forms of iron in vivo, mechanisms involved in iron uptake and release and their regulation. Furthermore, the model is interfaced with drug pharmacokinetics to allow simulation of treatment dynamics. We calibrated our model with experimental and clinical data from peer-reviewed literature to reliably simulate CKD anemia and the effects of current treatment involving combination of epoietin-alpha and iron dextran. This in silico whole-body model of iron metabolism predicts that a year of treatment can potentially lead to 90% downregulation of ferroportin (FPN) levels, 15-fold increase in iron stores with only a 20% increase in iron flux from the reticulo-endothelial system (RES). Model simulations quantified unmeasured iron fluxes, previously unknown effects of treatment on FPN-level and iron stores in the RES. This mechanistic whole-body model can be the basis for future studies that incorporate iron metabolism together with related clinical experiments. Such an approach could pave the way for development of effective personalized treatment of CKD anemia.


Asunto(s)
Anemia/metabolismo , Anemia/terapia , Hierro/metabolismo , Modelos Biológicos , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Anemia/etiología , Transporte Biológico Activo , Médula Ósea/metabolismo , Proteínas de Transporte de Catión/metabolismo , Biología Computacional , Epoetina alfa/uso terapéutico , Eritrocitos/metabolismo , Eritropoyetina/metabolismo , Hepcidinas/metabolismo , Humanos , Hierro/sangre , Complejo Hierro-Dextran/uso terapéutico , Hígado/metabolismo , Sistema Mononuclear Fagocítico/metabolismo , Transferrina/metabolismo
8.
Gastroenterology ; 153(1): 219-232, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28400196

RESUMEN

BACKGROUND & AIMS: Variants in the tumor necrosis factor superfamily member 15 gene (TNFSF15, also called TL1A) have been associated with risk for inflammatory bowel disease (IBD). TL1A affects expression of multiple cytokines to promote mucosal inflammation. Little is known about the TL1A-response pathways that regulate cytokine expression. We investigated T-cell gene expression patterns to determine the mechanisms by which TL1A regulates cytokine production, and whether these associate with outcomes of patients with Crohn's disease (CD). METHODS: Peripheral T cells isolated from normal donors were cultured with TL1A. We performed gene expression profile analysis by RNA sequencing of subsets of interferon gamma (IFNG)-producing and non-producing cells purified by flow cytometry. Unsupervised hierarchical clustering analysis was used to identify gene expression differences between these subsets. Ribonuclease T2 gene (RNASET2) expression and methylation were assessed by quantitative trait loci analyses. Clinical characteristics of patients (complications, resistance to therapy, and recurrence time) were associated with single nucleotide polymorphisms in RNASET2. We performed motif screening to identify polymorphisms that disrupt transcription factor binding sites. Levels of RNASET2 were knocked down with small interfering RNA in CD4+ T cells and the effect on protein expression was determined by proteomic analysis and cytokine production. Cell aggregation was measured by flow cytometry. RESULTS: We identified 764 genes with at least a 2-fold difference in TL1A-mediated expression between IFNG-secreting and non-secreting T cells (P < 1 × 10-5). Many of these genes were located near IBD susceptibility variants. RNASET2 was the only IBD risk-associated gene with >5-fold down-regulation in the IFNG-secreting subset. RNASET2 disease risk variants were associated with decreased expression in peripheral and mucosal tissues and DNA hypermethylation in CD patients requiring surgical intervention. RNASET2 disease risk variants were associated in CD patients with more complicated disease or resistance to therapy, defined in part by failed response to treatment, increased length of intestinal resection, shorter time to repeat surgery, and high Rutgeerts score (>2) in postoperative endoscopy. The RNASET2 variant rs2149092 was predicted to disrupt a consensus binding site for the transcription factor ETS within an enhancer region. Expression of RNASET2 correlated with expression of ETS. RNASET2 knockdown in T cells increased expression of IFNG and intercellular adhesion molecule 1 (ICAM1) and induced T-cell aggregation. A blocking antibody against (ILFA1), disrupting the lymphocyte function-associated antigen 1-intercellular adhesion molecule 1 interaction, reduced T-cell production of IFNG. CONCLUSIONS: We identified decreased expression of RNASET2 as a component of TL1A-mediated increase in production of IFNG and as a potential biomarker for patients with severe CD. Further study of the role of RNASET2 in regulating mucosal inflammation may lead to development of novel therapeutic targets.


Asunto(s)
Enfermedad de Crohn/genética , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/metabolismo , Ribonucleasas/genética , Linfocitos T/efectos de los fármacos , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Proteínas Supresoras de Tumor/genética , Alelos , Agregación Celular , Células Cultivadas , Enfermedad de Crohn/cirugía , Metilación de ADN , Regulación hacia Abajo , Silenciador del Gen , Humanos , Molécula 1 de Adhesión Intercelular/genética , Mucosa Intestinal/metabolismo , Antígeno-1 Asociado a Función de Linfocito/inmunología , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-ets/genética , Índice de Severidad de la Enfermedad , Linfocitos T/metabolismo , Transcriptoma
9.
PLoS Biol ; 11(8): e1001635, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23976881

RESUMEN

Cell regulatory circuits integrate diverse, and sometimes conflicting, environmental cues to generate appropriate, condition-dependent responses. Here, we elucidate the components and mechanisms driving a protein-directed RNA switch in the 3'UTR of vascular endothelial growth factor (VEGF)-A. We describe a novel HILDA (hypoxia-inducible hnRNP L-DRBP76-hnRNP A2/B1) complex that coordinates a three-element RNA switch, enabling VEGFA mRNA translation during combined hypoxia and inflammation. In addition to binding the CA-rich element (CARE), heterogeneous nuclear ribonucleoprotein (hnRNP) L regulates switch assembly and function. hnRNP L undergoes two previously unrecognized, condition-dependent posttranslational modifications: IFN-γ induces prolyl hydroxylation and von Hippel-Lindau (VHL)-mediated proteasomal degradation, whereas hypoxia stimulates hnRNP L phosphorylation at Tyr(359), inducing binding to hnRNP A2/B1, which stabilizes the protein. Also, phospho-hnRNP L recruits DRBP76 (double-stranded RNA binding protein 76) to the 3'UTR, where it binds an adjacent AU-rich stem-loop (AUSL) element, "flipping" the RNA switch by disrupting the GAIT (interferon-gamma-activated inhibitor of translation) element, preventing GAIT complex binding, and driving robust VEGFA mRNA translation. The signal-dependent, HILDA complex coordinates the function of a trio of neighboring RNA elements, thereby regulating translation of VEGFA and potentially other mRNA targets. The VEGFA RNA switch might function to ensure appropriate angiogenesis and tissue oxygenation during conflicting signals from combined inflammation and hypoxia. We propose the VEGFA RNA switch as an archetype for signal-activated, protein-directed, multi-element RNA switches that regulate posttranscriptional gene expression in complex environments.


Asunto(s)
Regiones no Traducidas 3'/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , Proteínas del Factor Nuclear 90/metabolismo , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/genética , Línea Celular , Técnica del Anticuerpo Fluorescente , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo L/genética , Humanos , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Proteínas del Factor Nuclear 90/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitinación/genética , Ubiquitinación/fisiología
10.
PLoS Comput Biol ; 10(7): e1003701, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24991925

RESUMEN

A major process of iron homeostasis in whole-body iron metabolism is the release of iron from the macrophages of the reticuloendothelial system. Macrophages recognize and phagocytose senescent or damaged erythrocytes. Then, they process the heme iron, which is returned to the circulation for reutilization by red blood cell precursors during erythropoiesis. The amount of iron released, compared to the amount shunted for storage as ferritin, is greater during iron deficiency. A currently accepted model of iron release assumes a passive-gradient with free diffusion of intracellular labile iron (Fe2+) through ferroportin (FPN), the transporter on the plasma membrane. Outside the cell, a multi-copper ferroxidase, ceruloplasmin (Cp), oxidizes ferrous to ferric ion. Apo-transferrin (Tf), the primary carrier of soluble iron in the plasma, binds ferric ion to form mono-ferric and di-ferric transferrin. According to the passive-gradient model, the removal of ferrous ion from the site of release sustains the gradient that maintains the iron release. Subcellular localization of FPN, however, indicates that the role of FPN may be more complex. By experiments and mathematical modeling, we have investigated the detailed mechanism of iron release from macrophages focusing on the roles of the Cp, FPN and apo-Tf. The passive-gradient model is quantitatively analyzed using a mathematical model for the first time. A comparison of experimental data with model simulations shows that the passive-gradient model cannot explain macrophage iron release. However, a facilitated-transport model associated with FPN can explain the iron release mechanism. According to the facilitated-transport model, intracellular FPN carries labile iron to the macrophage membrane. Extracellular Cp accelerates the oxidation of ferrous ion bound to FPN. Apo-Tf in the extracellular environment binds to the oxidized ferrous ion, completing the release process. Facilitated-transport model can correctly predict cellular iron efflux and is essential for physiologically relevant whole-body model of iron metabolism.


Asunto(s)
Simulación por Computador , Homeostasis/fisiología , Hierro/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , Biología Computacional , Humanos , Espacio Intracelular/metabolismo , Macrófagos/citología
11.
Inflamm Bowel Dis ; 29(10): 1622-1632, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37262302

RESUMEN

BACKGROUND: Sex is an integral variable often overlooked in complex disease genetics. Differences between sexes have been reported in natural history, disease complications, and age of onset in inflammatory bowel disease (IBD). While association studies have identified >230 IBD loci, there have been a limited number of studies investigating sex differences underlying these genetic associations. METHODS: We report the first investigation of sex-dimorphic associations via meta-analysis of a sex-stratified association study (34 579 IBD cases, 39 125 controls). In addition, we performed chromosome (chr) X-specific analyses, considering models of X inactivation (XCI) and XCI escape. Demographic and clinical characteristics were also compared between sexes. RESULTS: We identified significant differences between sexes for disease location and perianal complication in Crohn's disease and disease extent in ulcerative colitis. We observed genome-wide-significant sex-dimorphic associations (P < 5 × 10-8) at loci not previously reported in large-scale IBD genetic studies, including at chr9q22, CARMIL1, and UBASH3A. We identified variants in known IBD loci, including in chr2p15 and within the major histocompatibility complex on chr6, exhibiting sex-specific patterns of association (P < 5 × 10-7 in one sex only). We identified 3 chrX associations with IBD, including a novel Crohn's disease susceptibility locus at Xp22. CONCLUSIONS: These analyses identified novel IBD loci, in addition to characterizing sex-specific patterns of associations underlying sex-dimorphic associations. By elucidating the role of sex in IBD genetics, our study will help enhance our understanding of the differences between the sexes in IBD biology and underscores a need to move beyond conventional sex-combined analyses to appreciate the genetic architecture of IBD more comprehensively.


Sex-dimorphic meta-analyses of sex-stratified case-control (n = 73 704) regression identified 3 novel inflammatory bowel disease loci reaching genome-wide significance and highlighted chromosome 2 and major histocompatibility complex variants exhibiting sex-specific association. In addition, a novel chromosome X Crohn's disease susceptibility locus was identified.


Asunto(s)
Colitis Ulcerosa , Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Humanos , Femenino , Masculino , Predisposición Genética a la Enfermedad , Enfermedades Inflamatorias del Intestino/genética , Enfermedad de Crohn/genética , Colitis Ulcerosa/genética , Caracteres Sexuales , Estudio de Asociación del Genoma Completo
12.
Prostate ; 71(2): 184-96, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20672321

RESUMEN

BACKGROUND: Matriptase, a type II transmembrane serine protease, has been linked to initiation and promotion of epidermal carcinogenesis in a murine model, suggesting that deregulation of its role in epithelia contributes to transformation. In human prostate cancer, matriptase expression correlates with progression. It is therefore of interest to determine how matriptase may contribute to epithelial neoplastic progression. One approach for studying this is to identify potential matriptase substrates involved in epithelial integrity and/or transformation like the extracellular matrix macromolecule, laminin-332 (Ln-332), which is found in the basement membrane of many epithelia, including prostate. Proteolytic processing of Ln-332 regulates cell motility of both normal and transformed cells, which has implications in cancer progression. METHODS: In vitro cleavage experiments were performed with purified Ln-332 protein and matriptase. Western blotting, enzyme inhibition assays, and mass spectrometry were used to confirm cleavage events. Matriptase overexpressing LNCaP prostate cancer cells were generated and included in Transwell migration assays and single cell motility assays, along with other prostate cells. RESULTS: We report that matriptase proteolytically cleaves Ln-332 in the ß3 chain. Substrate specificity was confirmed by blocking cleavage with the matriptase inhibitor, Kunitz domain-1. Transwell migration assays showed that DU145 cell motility was significantly enhanced when plated on matriptase-cleaved Ln-332. Similarly, Transwell migration of matriptase-overexpressing LNCaP cells was significantly increased on Ln-332 and, as determined by live single-cell microscopy, two motility parameters of this cell line, speed and directional persistence, were also higher. CONCLUSIONS: Proteolytic processing of Ln-332 by matriptase enhances speed and directional persistence of prostate cancer cells.


Asunto(s)
Movimiento Celular/fisiología , Laminina/metabolismo , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/metabolismo , Línea Celular Tumoral , Humanos , Masculino , Microscopía de Contraste de Fase , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Inhibidoras de Proteinasas Secretoras/farmacología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Cancer Cell ; 39(9): 1202-1213.e6, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34329585

RESUMEN

Studies suggest that the efficacy of cancer chemotherapy and immunotherapy is influenced by intestinal bacteria. However, the influence of the microbiome on radiation therapy is not as well understood, and the microbiome comprises more than bacteria. Here, we find that intestinal fungi regulate antitumor immune responses following radiation in mouse models of breast cancer and melanoma and that fungi and bacteria have opposite influences on these responses. Antibiotic-mediated depletion or gnotobiotic exclusion of fungi enhances responsiveness to radiation, whereas antibiotic-mediated depletion of bacteria reduces responsiveness and is associated with overgrowth of commensal fungi. Further, elevated intratumoral expression of Dectin-1, a primary innate sensor of fungi, is negatively associated with survival in patients with breast cancer and is required for the effects of commensal fungi in mouse models of radiation therapy.


Asunto(s)
Antifúngicos/administración & dosificación , Bacterias/clasificación , Neoplasias de la Mama/terapia , Hongos/efectos de los fármacos , Lectinas Tipo C/genética , Melanoma/terapia , Animales , Antifúngicos/farmacología , Bacterias/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/microbiología , Terapia Combinada , Regulación hacia Abajo , Femenino , Hongos/clasificación , Hongos/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Melanoma/inmunología , Melanoma/microbiología , Ratones , Simbiosis , Linfocitos T/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Biophys J ; 99(2): 367-76, 2010 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-20643054

RESUMEN

Cell motility is a fundamental process with relevance to embryonic development, immune response, and metastasis. Cells move either spontaneously, in a nondirected fashion, or in response to chemotactic signals, in a directed fashion. Even though they are often studied separately, both forms of motility share many complex processes at the molecular and subcellular scale, e.g., orchestrated cytoskeletal rearrangements and polarization. In addition, at the cellular level both types of motility include persistent runs interspersed with reorientation pauses. Because there is a great range of variability in motility among different cell types, a key challenge in the field is to integrate these multiscale processes into a coherent framework. We analyzed the motility of Dictyostelium cells with bimodal analysis, a method that compares time spent in persistent versus reorientation mode. Unexpectedly, we found that reorientation time is coupled with persistent time in an inverse correlation and, surprisingly, the inverse correlation holds for both nondirected and chemotactic motility, so that the full range of Dictyostelium motility can be described by a single scaling relationship. Additionally, we found an identical scaling relationship for three human cell lines, indicating that the coupling of reorientation and persistence holds across species and making it possible to describe the complexity of cell motility in a surprisingly general and simple manner. With this new perspective, we analyzed the motility of Dictyostelium mutants, and found four in which the coupling between two modes was altered. Our results point to a fundamental underlying principle, described by a simple scaling law, unifying mechanisms of eukaryotic cell motility at several scales.


Asunto(s)
Quimiotaxis , Dictyostelium/citología , Modelos Biológicos , Línea Celular , Dictyostelium/metabolismo , Células Eucariotas/citología , Células Eucariotas/metabolismo , Humanos , Mutación/genética , Factores de Tiempo
15.
medRxiv ; 2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32511625

RESUMEN

Angiotensin-Converting Enzyme 2 ( ACE2 ) has been identified as the host receptor for SARS-coronavirus 2 (SARS-CoV-2) which has infected millions world-wide and likely caused hundreds of thousands of deaths. Utilizing transcriptomic data from four cohorts taken from Crohn's disease (CD) and non-inflammatory bowel disease (IBD) subjects, we observed evidence of increased ACE2 mRNA in ileum with demographic features that have been associated with poor outcomes in COVID-19 including age and raised BMI. ACE2 was downregulated in CD compared to controls in independent cohorts. Within CD, ACE2 expression was reduced in inflamed ileal tissue and also remarkably, from uninvolved tissue in patients with a worse prognosis in both adult and pediatric cohorts. In active CD, small bowel ACE2 expression was restored by anti-TNF therapy particularly in anti-TNF responders. Collectively our data suggest that ACE2 downregulation is associated with inflammation and worse outcomes in CD.

16.
J Crohns Colitis ; 13(8): 1055-1066, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-30877309

RESUMEN

BACKGROUND AND AIMS: Heterogeneity in Crohn's disease [CD] provides a challenge for the development of effective therapies. Our goal was to define a unique molecular signature for severe, refractory CD to enable precision therapy approaches to disease treatment and to facilitate earlier intervention in complicated disease. METHODS: We analysed clinical metadata, genetics, and transcriptomics from uninvolved ileal tissue from CD patients who underwent a single small bowel resection. We determined transcriptional risk scores, cellular signatures, and mechanistic pathways that define patient subsets in refractory CD. RESULTS: Within refractory CD, we found three CD patient subgroups [CD1, CD2, and CD3]. Compared with CD1, CD3 was enriched for subjects with increased disease recurrence after first surgery [OR = 6.78, p = 0.04], enhanced occurrence of second surgery [OR = 5.07, p = 0.016], and presence of perianal CD [OR = 3.61, p = 0.036]. The proportion of patients with recurrence-free survival was smaller in CD3 than in CD1 (p = 0.02, median survival time [months] in CD1 = 10 and CD3 = 6). Overlaying differential gene expression between CD1 and CD3 on CD subgroup-associated genetic polymorphisms identified 174 genes representing both genetic and biological differences between the CD subgroups. Pathway analyses using this unique gene signature indicated eukaryotic initiation factor 2 [eIF2] and cyclic adenosine monophosphate [cAMP] signalling to be dominant pathways associated with CD3. Furthermore, the severe, refractory subset, CD3, was associated with a higher transcriptional risk score and enriched with eosinophil and natural killer T [NKT] cell gene signatures. CONCLUSION: We characterized a subset of severe, refractory CD patients who may need more aggressive treatment after first resection and who are likely to benefit from targeted therapy based on their genotype and tissue gene expression signature.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedad de Crohn , Íleon , Pruebas de Farmacogenómica , Transducción de Señal/genética , Adolescente , Adulto , Niño , Enfermedad de Crohn/diagnóstico , Enfermedad de Crohn/epidemiología , Enfermedad de Crohn/genética , Enfermedad de Crohn/terapia , Procedimientos Quirúrgicos del Sistema Digestivo/métodos , Femenino , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/estadística & datos numéricos , Estudio de Asociación del Genoma Completo , Humanos , Íleon/metabolismo , Íleon/patología , Íleon/cirugía , Masculino , Selección de Paciente , Índice de Severidad de la Enfermedad , Estados Unidos
17.
Mucosal Immunol ; 12(3): 644-655, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30617301

RESUMEN

T helper 9 (TH9) cells are important for the development of inflammatory and allergic diseases. The TH9 transcriptional network converges signals from cytokines and antigen presentation but is incompletely understood. Here, we identified TL1A, a member of the TNF superfamily, as a strong inducer of mouse and human TH9 differentiation. Mechanistically, TL1A induced the expression of the transcription factors BATF and BATF3 and facilitated their binding to the Il9 promoter leading to enhanced secretion of IL-9. BATF- and BATF3-deficiencies impaired IL-9 secretion under TH9 and TH9-TL1A-polarizing conditions. In vivo, using a T-cell transfer model, we demonstrated that TL1A promoted IL-9-dependent, TH9 cell-induced intestinal and lung inflammation. Neutralizing IL-9 antibodies attenuated TL1A-driven mucosal inflammation. Batf3-/- TH9-TL1A cells induced reduced inflammation and cytokine expression in vivo compared to WT cells. Our results demonstrate that TL1A promotes TH9 cell differentiation and function and define a role for BATF3 in T-cell-driven mucosal inflammation.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-9/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Neutralizantes/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Diferenciación Celular , Células Cultivadas , Humanos , Interleucina-9/genética , Interleucina-9/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Represoras/genética , Transducción de Señal , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
18.
Inflamm Bowel Dis ; 24(11): 2413-2422, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29860388

RESUMEN

Background: Age of onset is linked to variations in clinical phenotypes and natural history in Crohn's disease (CD). We aim to define etiologically more homogenous subgroups in CD based on ages of onset. Methods: We examined the distribution of CD polygenetic risk score (PRS) across ages of diagnosis in a Caucasian cohort of 2344 independent CD patients. We identified subgroups with a distinct distribution of PRS and compared those groups in genetics, demographic characteristics, clinical subphenotypes, and serological markers. The results were replicated in an independent cohort of 13,065 CD patients from the International Inflammatory Bowel Diseases Genetic Consortium (IIBDGC). Results: We identified a late-onset (LO) subgroup in CD (age at diagnosis ≥ 55 years) with significantly lower PRS compared with the intermediate group (age at diagnosis between 5 and 55 years) in both cohorts. Smoking cessation, a risk factor for ulcerative colitis (UC) and protective factor for CD, had a higher rate in this LO subgroup in comparison with the intermediate group. We also compared the LO group with the intermediate group, and, consistent with previous reports, the LO group more often had colonic CD, had less penetrating disease behavior, and had less need for surgery. Serological analysis showed that LO CD patients were more antineutrophil cytoplasmic antibody positive and less antisaccharomyces cerevisiae antibody positive compared with the intermediate group. Variance component analysis indicated that overall genetic contribution to LO CD was lower relative to the middle group, and genetic heterogeneity testing indicated that LO CD was different from the middle group in underlying genetic architecture. Conclusions: Late-onset CD is subgroup distinct in genetic and behavioral risk factors with UC-like characteristics. 10.1093/ibd/izy148_video1izy148.video15791413461001.


Asunto(s)
Colitis Ulcerosa/patología , Enfermedad de Crohn/clasificación , Enfermedad de Crohn/patología , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Colitis Ulcerosa/epidemiología , Colitis Ulcerosa/genética , Enfermedad de Crohn/epidemiología , Enfermedad de Crohn/genética , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Factores de Riesgo , Estados Unidos/epidemiología , Adulto Joven
19.
Nat Cell Biol ; 16(5): 445-56, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24747440

RESUMEN

The tumour vascular microenvironment supports tumorigenesis not only by supplying oxygen and diffusible nutrients but also by secreting soluble factors that promote tumorigenesis. Here we identify a feedforward mechanism in which endothelial cells (ECs), in response to tumour-derived mediators, release angiocrines driving aberrant vascularization and glioblastoma multiforme (GBM) progression through a hypoxia-independent induction of hypoxia-inducible factor (HIF)-1α. Phosphorylation of profilin-1 (Pfn-1) at Tyr 129 in ECs induces binding to the tumour suppressor protein von Hippel-Lindau (VHL), and prevents VHL-mediated degradation of prolyl-hydroxylated HIF-1α, culminating in HIF-1α accumulation even in normoxia. Elevated HIF-1α induces expression of multiple angiogenic factors, leading to vascular abnormality and tumour progression. In a genetic model of GBM, mice with an EC-specific defect in Pfn-1 phosphorylation exhibit reduced tumour angiogenesis, normalized vasculature and improved survival. Moreover, EC-specific Pfn-1 phosphorylation is associated with tumour aggressiveness in human glioma. These findings suggest that targeting Pfn-1 phosphorylation may offer a selective strategy for therapeutic intervention of malignant solid tumours.


Asunto(s)
Comunicación Autocrina , Neoplasias Encefálicas/metabolismo , Células Endoteliales/metabolismo , Glioblastoma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Profilinas/metabolismo , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Permeabilidad Capilar , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/patología , Femenino , Glioblastoma/irrigación sanguínea , Glioblastoma/genética , Glioblastoma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Clasificación del Tumor , Neovascularización Patológica , Fosforilación , Profilinas/genética , Interferencia de ARN , Factores de Tiempo , Transfección , Carga Tumoral , Microambiente Tumoral , Tirosina , Regulación hacia Arriba , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
20.
PLoS One ; 5(3): e9636, 2010 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-20224792

RESUMEN

BACKGROUND: Organisms, at scales ranging from unicellular to mammals, have been known to exhibit foraging behavior described by random walks whose segments confirm to Lévy or exponential distributions. For the first time, we present evidence that single cells (mammary epithelial cells) that exist in multi-cellular organisms (humans) follow a bimodal correlated random walk (BCRW). METHODOLOGY/PRINCIPAL FINDINGS: Cellular tracks of MCF-10A pBabe, neuN and neuT random migration on 2-D plastic substrates, analyzed using bimodal analysis, were found to reveal the BCRW pattern. We find two types of exponentially distributed correlated flights (corresponding to what we refer to as the directional and re-orientation phases) each having its own correlation between move step-lengths within flights. The exponential distribution of flight lengths was confirmed using different analysis methods (logarithmic binning with normalization, survival frequency plots and maximum likelihood estimation). CONCLUSIONS/SIGNIFICANCE: Because of the presence of non-uniform turn angle distribution of move step-lengths within a flight and two different types of flights, we propose that the epithelial random walk is a BCRW comprising of two alternating modes with varying degree of correlations, rather than a simple persistent random walk. A BCRW model rather than a simple persistent random walk correctly matches the super-diffusivity in the cell migration paths as indicated by simulations based on the BCRW model.


Asunto(s)
Movimiento Celular , Células Epiteliales/patología , Glándulas Mamarias Humanas/patología , Algoritmos , Simulación por Computador , Difusión , Células Epiteliales/fisiología , Humanos , Funciones de Verosimilitud , Glándulas Mamarias Humanas/fisiología , Modelos Biológicos , Modelos Estadísticos , Modelos Teóricos , Distribución Normal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA