Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(22): 4099-4116.e13, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36261039

RESUMEN

Some people are more attractive to mosquitoes than others, but the mechanistic basis of this phenomenon is poorly understood. We tested mosquito attraction to human skin odor and identified people who are exceptionally attractive or unattractive to mosquitoes. These differences were stable over several years. Chemical analysis revealed that highly attractive people produce significantly more carboxylic acids in their skin emanations. Mutant mosquitoes lacking the chemosensory co-receptors Ir8a, Ir25a, or Ir76b were severely impaired in attraction to human scent, but retained the ability to differentiate highly and weakly attractive people. The link between elevated carboxylic acids in "mosquito-magnet" human skin odor and phenotypes of genetic mutations in carboxylic acid receptors suggests that such compounds contribute to differential mosquito attraction. Understanding why some humans are more attractive than others provides insights into what skin odorants are most important to the mosquito and could inform the development of more effective repellents.


Asunto(s)
Aedes , Anopheles , Repelentes de Insectos , Animales , Humanos , Ácidos Carboxílicos/farmacología , Odorantes/análisis , Repelentes de Insectos/farmacología , Repelentes de Insectos/análisis
2.
Nat Immunol ; 24(8): 1358-1369, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37365386

RESUMEN

Following infection or vaccination, activated B cells at extrafollicular sites or within germinal centers (GCs) undergo vigorous clonal proliferation. Proliferating lymphocytes have been shown to undertake lactate dehydrogenase A (LDHA)-dependent aerobic glycolysis; however, the specific role of this metabolic pathway in a B cell transitioning from a naïve to a highly proliferative, activated state remains poorly defined. Here, we deleted LDHA in a stage-specific and cell-specific manner. We find that ablation of LDHA in a naïve B cell did not profoundly affect its ability to undergo a bacterial lipopolysaccharide-induced extrafollicular B cell response. On the other hand, LDHA-deleted naïve B cells had a severe defect in their capacities to form GCs and mount GC-dependent antibody responses. In addition, loss of LDHA in T cells severely compromised B cell-dependent immune responses. Strikingly, when LDHA was deleted in activated, as opposed to naïve, B cells, there were only minimal effects on the GC reaction and in the generation of high-affinity antibodies. These findings strongly suggest that naïve and activated B cells have distinct metabolic requirements that are further regulated by niche and cellular interactions.


Asunto(s)
Linfocitos B , Centro Germinal , Linfocitos T , Activación de Linfocitos , Comunicación Celular
3.
Nat Immunol ; 21(9): 1022-1033, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661364

RESUMEN

The majority of tumor-infiltrating T cells exhibit a terminally exhausted phenotype, marked by a loss of self-renewal capacity. How repetitive antigenic stimulation impairs T cell self-renewal remains poorly defined. Here, we show that persistent antigenic stimulation impaired ADP-coupled oxidative phosphorylation. The resultant bioenergetic compromise blocked proliferation by limiting nucleotide triphosphate synthesis. Inhibition of mitochondrial oxidative phosphorylation in activated T cells was sufficient to suppress proliferation and upregulate genes linked to T cell exhaustion. Conversely, prevention of mitochondrial oxidative stress during chronic T cell stimulation allowed sustained T cell proliferation and induced genes associated with stem-like progenitor T cells. As a result, antioxidant treatment enhanced the anti-tumor efficacy of chronically stimulated T cells. These data reveal that loss of ATP production through oxidative phosphorylation limits T cell proliferation and effector function during chronic antigenic stimulation. Furthermore, treatments that maintain redox balance promote T cell self-renewal and enhance anti-tumor immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Mitocondrias/metabolismo , Neoplasias/inmunología , Adenosina Difosfato/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Antioxidantes/farmacología , Proliferación Celular , Autorrenovación de las Células , Anergia Clonal/genética , Metabolismo Energético , Tolerancia Inmunológica , Activación de Linfocitos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Fosforilación Oxidativa
4.
Nat Immunol ; 17(6): 656-65, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27043409

RESUMEN

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.


Asunto(s)
Arginasa/metabolismo , Linfocitos/fisiología , Neumonía/inmunología , Animales , Arginasa/genética , Proliferación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Glucólisis/genética , Humanos , Inmunidad Innata , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Poliaminas/metabolismo , Células Th2/inmunología
5.
Nat Immunol ; 16(8): 838-849, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26147685

RESUMEN

Interferon-γ (IFN-γ) primes macrophages for enhanced microbial killing and inflammatory activation by Toll-like receptors (TLRs), but little is known about the regulation of cell metabolism or mRNA translation during this priming. We found that IFN-γ regulated the metabolism and mRNA translation of human macrophages by targeting the kinases mTORC1 and MNK, both of which converge on the selective regulator of translation initiation eIF4E. Physiological downregulation of mTORC1 by IFN-γ was associated with autophagy and translational suppression of repressors of inflammation such as HES1. Genome-wide ribosome profiling in TLR2-stimulated macrophages showed that IFN-γ selectively modulated the macrophage translatome to promote inflammation, further reprogram metabolic pathways and modulate protein synthesis. These results show that IFN-γ-mediated metabolic reprogramming and translational regulation are key components of classical inflammatory macrophage activation.


Asunto(s)
Interferón gamma/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Biosíntesis de Proteínas/inmunología , ARN Mensajero/inmunología , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Células Cultivadas , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/inmunología , Factor 4E Eucariótico de Iniciación/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Proteínas de Homeodominio/metabolismo , Humanos , Interferón gamma/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , MicroARNs/genética , Microscopía Fluorescente , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Factor de Transcripción HES-1
6.
Nature ; 581(7809): 475-479, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32461639

RESUMEN

Intestinal health relies on the immunosuppressive activity of CD4+ regulatory T (Treg) cells1. Expression of the transcription factor Foxp3 defines this lineage, and can be induced extrathymically by dietary or commensal-derived antigens in a process assisted by a Foxp3 enhancer known as conserved non-coding sequence 1 (CNS1)2-4. Products of microbial fermentation including butyrate facilitate the generation of peripherally induced Treg (pTreg) cells5-7, indicating that metabolites shape the composition of the colonic immune cell population. In addition to dietary components, bacteria modify host-derived molecules, generating a number of biologically active substances. This is epitomized by the bacterial transformation of bile acids, which creates a complex pool of steroids8 with a range of physiological functions9. Here we screened the major species of deconjugated bile acids for their ability to potentiate the differentiation of pTreg cells. We found that the secondary bile acid 3ß-hydroxydeoxycholic acid (isoDCA) increased Foxp3 induction by acting on dendritic cells (DCs) to diminish their immunostimulatory properties. Ablating one receptor, the farnesoid X receptor, in DCs enhanced the generation of Treg cells and imposed a transcriptional profile similar to that induced by isoDCA, suggesting an interaction between this bile acid and nuclear receptor. To investigate isoDCA in vivo, we took a synthetic biology approach and designed minimal microbial consortia containing engineered Bacteroides strains. IsoDCA-producing consortia increased the number of colonic RORγt-expressing Treg cells in a CNS1-dependent manner, suggesting enhanced extrathymic differentiation.


Asunto(s)
Bacterias/metabolismo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Bacteroides/metabolismo , Colon/microbiología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Fermentación , Microbioma Gastrointestinal , Masculino , Ratones , Ratones Endogámicos C57BL , Consorcios Microbianos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
8.
Nature ; 583(7816): 441-446, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32641826

RESUMEN

Connections between the gut and brain monitor the intestinal tissue and its microbial and dietary content1, regulating both physiological intestinal functions such as nutrient absorption and motility2,3, and brain-wired feeding behaviour2. It is therefore plausible that circuits exist to detect gut microorganisms and relay this information to areas of the central nervous system that, in turn, regulate gut physiology4. Here we characterize the influence of the microbiota on enteric-associated neurons by combining gnotobiotic mouse models with transcriptomics, circuit-tracing methods and functional manipulations. We find that the gut microbiome modulates gut-extrinsic sympathetic neurons: microbiota depletion leads to increased expression of the neuronal transcription factor cFos, and colonization of germ-free mice with bacteria that produce short-chain fatty acids suppresses cFos expression in the gut sympathetic ganglia. Chemogenetic manipulations, translational profiling and anterograde tracing identify a subset of distal intestine-projecting vagal neurons that are positioned to have an afferent role in microbiota-mediated modulation of gut sympathetic neurons. Retrograde polysynaptic neuronal tracing from the intestinal wall identifies brainstem sensory nuclei that are activated during microbial depletion, as well as efferent sympathetic premotor glutamatergic neurons that regulate gastrointestinal transit. These results reveal microbiota-dependent control of gut-extrinsic sympathetic activation through a gut-brain circuit.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Intestinos/inervación , Neuronas/fisiología , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/fisiología , Animales , Disbiosis/fisiopatología , Femenino , Ganglios Simpáticos/citología , Ganglios Simpáticos/fisiología , Motilidad Gastrointestinal , Vida Libre de Gérmenes , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Vías Nerviosas/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transcriptoma
9.
Nat Chem Biol ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37884806

RESUMEN

Impaired redox metabolism is a key contributor to the etiology of many diseases, including primary mitochondrial disorders, cancer, neurodegeneration and aging. However, mechanistic studies of redox imbalance remain challenging due to limited strategies that can perturb redox metabolism in various cellular or organismal backgrounds. Most studies involving impaired redox metabolism have focused on oxidative stress; consequently, less is known about the settings where there is an overabundance of NADH reducing equivalents, termed reductive stress. Here we introduce a soluble transhydrogenase from Escherichia coli (EcSTH) as a novel genetically encoded tool to promote reductive stress in living cells. When expressed in mammalian cells, EcSTH, and a mitochondrially targeted version (mitoEcSTH), robustly elevated the NADH/NAD+ ratio in a compartment-specific manner. Using this tool, we determined that metabolic and transcriptomic signatures of the NADH reductive stress are cellular background specific. Collectively, our novel genetically encoded tool represents an orthogonal strategy to promote reductive stress.

10.
Nature ; 572(7771): 665-669, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31435014

RESUMEN

Intestinal commensal bacteria can inhibit dense colonization of the gut by vancomycin-resistant Enterococcus faecium (VRE), a leading cause of hospital-acquired infections1,2. A four-strained consortium of commensal bacteria that contains Blautia producta BPSCSK can reverse antibiotic-induced susceptibility to VRE infection3. Here we show that BPSCSK reduces growth of VRE by secreting a lantibiotic that is similar to the nisin-A produced by Lactococcus lactis. Although the growth of VRE is inhibited by BPSCSK and L. lactis in vitro, only BPSCSK colonizes the colon and reduces VRE density in vivo. In comparison to nisin-A, the BPSCSK lantibiotic has reduced activity against intestinal commensal bacteria. In patients at high risk of VRE infection, high abundance of the lantibiotic gene is associated with reduced density of E. faecium. In germ-free mice transplanted with patient-derived faeces, resistance to VRE colonization correlates with abundance of the lantibiotic gene. Lantibiotic-producing commensal strains of the gastrointestinal tract reduce colonization by VRE and represent potential probiotic agents to re-establish resistance to VRE.


Asunto(s)
Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Enterococcus faecium/efectos de los fármacos , Lactococcus lactis/metabolismo , Probióticos , Resistencia a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Animales , Antibacterianos/biosíntesis , Antibacterianos/aislamiento & purificación , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacteriocinas/genética , Bacteriocinas/aislamiento & purificación , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Heces/microbiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/crecimiento & desarrollo , Humanos , Lactococcus lactis/química , Lactococcus lactis/crecimiento & desarrollo , Lactococcus lactis/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Microbiota/genética , Nisina/química , Nisina/farmacología , Simbiosis/efectos de los fármacos , Vancomicina/farmacología , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
11.
EMBO J ; 39(8): e103334, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32134147

RESUMEN

The production and secretion of matrix proteins upon stimulation of fibroblasts by transforming growth factor-beta (TGFß) play a critical role in wound healing. How TGFß supports the bioenergetic cost of matrix protein synthesis is not fully understood. Here, we show that TGFß promotes protein translation at least in part by increasing the mitochondrial oxidation of glucose and glutamine carbons to support the bioenergetic demand of translation. Surprisingly, we found that in addition to stimulating the entry of glucose and glutamine carbon into the TCA cycle, TGFß induced the biosynthesis of proline from glutamine in a Smad4-dependent fashion. Metabolic manipulations that increased mitochondrial redox generation promoted proline biosynthesis, while reducing mitochondrial redox potential and/or ATP synthesis impaired proline biosynthesis. Thus, proline biosynthesis acts as a redox vent, preventing the TGFß-induced increase in mitochondrial glucose and glutamine catabolism from generating damaging reactive oxygen species (ROS) when TCA cycle activity exceeds the ability of oxidative phosphorylation to convert mitochondrial redox potential into ATP. In turn, the enhanced synthesis of proline supports TGFß-induced production of matrix proteins.


Asunto(s)
Fibrosis/metabolismo , Glucosa/metabolismo , Glutamina/metabolismo , Mitocondrias/metabolismo , Prolina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Ciclo del Ácido Cítrico , Colágeno/metabolismo , Metabolismo Energético , Humanos , Ratones , Células 3T3 NIH , Oxidación-Reducción , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo
12.
Blood ; 139(15): 2392-2405, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-34653248

RESUMEN

The intestinal microbiota is essential for the fermentation of dietary fiber into short-chain fatty acids (SCFA) such as butyrate, acetate, and propionate. SCFAs can bind to the G-protein-coupled receptors GPR43 and GPR109A (HCAR2), with varying affinities to promote cellular effects in metabolism or changes in immune function. We explored the role of GPR109A as the main receptor for butyrate in mouse models of allogeneic hematopoietic cell transplantation (allo-HCT) and graft-versus-host disease (GVHD). Deletion of GPR109A in allo-HCT recipients did not affect GVHD, but transplantation of T cells from GPR109A knockout (KO) (Gpr109a-/-) mice into allo-HCT recipient mice significantly reduced GVHD morbidity and mortality compared with recipients of wild-type (WT) T cells. Recipients of Gpr109a-/- T cells exhibited less GVHD-associated target organ pathology and decreased proliferation and homing of alloreactive T cells to target tissues. Although Gpr109a-/- T cells did not exhibit immune deficits at a steady state, following allo-activation, Gpr109a-/- T cells underwent increased apoptosis and were impaired mitochondrial oxidative phosphorylation, which was reversible through antioxidant treatment with N-acetylcysteine (NAC). In conclusion, we found that GPR109A expression by allo-activated T cells is essential for metabolic homeostasis and expansion, which are necessary features to induce GVHD after allo-HCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Butiratos , Ácidos Grasos Volátiles/fisiología , Ratones , Linfocitos T
13.
Nat Chem Biol ; 18(5): 565-574, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35165443

RESUMEN

Inflammasomes are multiprotein complexes that sense intracellular danger signals and induce pyroptosis. CARD8 and NLRP1 are related inflammasomes that are repressed by the enzymatic activities and protein structures of the dipeptidyl peptidases 8 and 9 (DPP8/9). Potent DPP8/9 inhibitors such as Val-boroPro (VbP) activate both NLRP1 and CARD8, but chemical probes that selectively activate only one have not been identified. Here we report a small molecule called CQ31 that selectively activates CARD8. CQ31 inhibits the M24B aminopeptidases prolidase (PEPD) and Xaa-Pro aminopeptidase 1 (XPNPEP1), leading to the accumulation of proline-containing peptides that inhibit DPP8/9 and thereby activate CARD8. NLRP1 is distinct from CARD8 in that it directly contacts DPP8/9's active site; these proline-containing peptides, unlike VbP, do not disrupt this repressive interaction and thus do not activate NLRP1. We expect that CQ31 will now become a valuable tool to study CARD8 biology.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Inflamasomas , Aminopeptidasas/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Neoplasias , Prolina
15.
Nature ; 559(7712): 125-129, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29950729

RESUMEN

Somatic mutations in the isocitrate dehydrogenase 2 gene (IDH2) contribute to the pathogenesis of acute myeloid leukaemia (AML) through the production of the oncometabolite 2-hydroxyglutarate (2HG)1-8. Enasidenib (AG-221) is an allosteric inhibitor that binds to the IDH2 dimer interface and blocks the production of 2HG by IDH2 mutants9,10. In a phase I/II clinical trial, enasidenib inhibited the production of 2HG and induced clinical responses in relapsed or refractory IDH2-mutant AML11. Here we describe two patients with IDH2-mutant AML who had a clinical response to enasidenib followed by clinical resistance, disease progression, and a recurrent increase in circulating levels of 2HG. We show that therapeutic resistance is associated with the emergence of second-site IDH2 mutations in trans, such that the resistance mutations occurred in the IDH2 allele without the neomorphic R140Q mutation. The in trans mutations occurred at glutamine 316 (Q316E) and isoleucine 319 (I319M), which are at the interface where enasidenib binds to the IDH2 dimer. The expression of either of these mutant disease alleles alone did not induce the production of 2HG; however, the expression of the Q316E or I319M mutation together with the R140Q mutation in trans allowed 2HG production that was resistant to inhibition by enasidenib. Biochemical studies predicted that resistance to allosteric IDH inhibitors could also occur via IDH dimer-interface mutations in cis, which was confirmed in a patient with acquired resistance to the IDH1 inhibitor ivosidenib (AG-120). Our observations uncover a mechanism of acquired resistance to a targeted therapy and underscore the importance of 2HG production in the pathogenesis of IDH-mutant malignancies.


Asunto(s)
Aminopiridinas/farmacología , Resistencia a Antineoplásicos/genética , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Leucemia Mieloide Aguda/genética , Proteínas Mutantes/genética , Mutación , Multimerización de Proteína/genética , Triazinas/farmacología , Alelos , Sitio Alostérico/efectos de los fármacos , Sitio Alostérico/genética , Aminopiridinas/química , Aminopiridinas/uso terapéutico , Animales , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Glutamina/genética , Glutaratos/sangre , Glutaratos/metabolismo , Células HEK293 , Humanos , Isoleucina/genética , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Mutantes/antagonistas & inhibidores , Triazinas/química , Triazinas/uso terapéutico
16.
J Inherit Metab Dis ; 46(5): 931-942, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37309295

RESUMEN

Toxicity of accumulating substrates is a significant problem in several disorders of valine and isoleucine degradation notably short-chain enoyl-CoA hydratase (ECHS1 or crotonase) deficiency, 3-hydroxyisobutyryl-CoA hydrolase (HIBCH) deficiency, propionic acidemia (PA), and methylmalonic aciduria (MMA). Isobutyryl-CoA dehydrogenase (ACAD8) and short/branched-chain acyl-CoA dehydrogenase (SBCAD, ACADSB) function in the valine and isoleucine degradation pathways, respectively. Deficiencies of these acyl-CoA dehydrogenase (ACAD) enzymes are considered biochemical abnormalities with limited or no clinical consequences. We investigated whether substrate reduction therapy through inhibition of ACAD8 and SBCAD can limit the accumulation of toxic metabolic intermediates in disorders of valine and isoleucine metabolism. Using analysis of acylcarnitine isomers, we show that 2-methylenecyclopropaneacetic acid (MCPA) inhibited SBCAD, isovaleryl-CoA dehydrogenase, short-chain acyl-CoA dehydrogenase and medium-chain acyl-CoA dehydrogenase, but not ACAD8. MCPA treatment of wild-type and PA HEK-293 cells caused a pronounced decrease in C3-carnitine. Furthermore, deletion of ACADSB in HEK-293 cells led to an equally strong decrease in C3-carnitine when compared to wild-type cells. Deletion of ECHS1 in HEK-293 cells caused a defect in lipoylation of the E2 component of the pyruvate dehydrogenase complex, which was not rescued by ACAD8 deletion. MCPA was able to rescue lipoylation in ECHS1 KO cells, but only in cells with prior ACAD8 deletion. SBCAD was not the sole ACAD responsible for this compensation, which indicates substantial promiscuity of ACADs in HEK-293 cells for the isobutyryl-CoA substrate. Substrate promiscuity appeared less prominent for 2-methylbutyryl-CoA at least in HEK-293 cells. We suggest that pharmacological inhibition of SBCAD to treat PA should be investigated further.


Asunto(s)
Ácido 2-Metil-4-clorofenoxiacético , Acidemia Propiónica , Humanos , Valina/genética , Valina/metabolismo , Acil-CoA Deshidrogenasa/metabolismo , Isoleucina/metabolismo , Células HEK293 , Carnitina
17.
Nature ; 549(7670): 48-53, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28854168

RESUMEN

Commensal bacteria are believed to have important roles in human health. The mechanisms by which they affect mammalian physiology remain poorly understood, but bacterial metabolites are likely to be key components of host interactions. Here we use bioinformatics and synthetic biology to mine the human microbiota for N-acyl amides that interact with G-protein-coupled receptors (GPCRs). We found that N-acyl amide synthase genes are enriched in gastrointestinal bacteria and the lipids that they encode interact with GPCRs that regulate gastrointestinal tract physiology. Mouse and cell-based models demonstrate that commensal GPR119 agonists regulate metabolic hormones and glucose homeostasis as efficiently as human ligands, although future studies are needed to define their potential physiological role in humans. Our results suggest that chemical mimicry of eukaryotic signalling molecules may be common among commensal bacteria and that manipulation of microbiota genes encoding metabolites that elicit host cellular responses represents a possible small-molecule therapeutic modality (microbiome-biosynthetic gene therapy).


Asunto(s)
Amidas/metabolismo , Bacterias/metabolismo , Mimetismo Biológico , Tracto Gastrointestinal/microbiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Simbiosis , Amidas/química , Animales , Bacterias/enzimología , Bacterias/genética , Glucemia/metabolismo , Femenino , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/metabolismo , Células HEK293 , Homeostasis , Humanos , Ligandos , Masculino , Ratones
18.
Mol Cell ; 58(2): 203-15, 2015 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-25818647

RESUMEN

Acetylation of histones at DNA regulatory elements plays a critical role in transcriptional activation. Histones are also modified by other acyl moieties, including crotonyl, yet the mechanisms that govern acetylation versus crotonylation and the functional consequences of this "choice" remain unclear. We show that the coactivator p300 has both crotonyltransferase and acetyltransferase activities, and that p300-catalyzed histone crotonylation directly stimulates transcription to a greater degree than histone acetylation. Levels of histone crotonylation are regulated by the cellular concentration of crotonyl-CoA, which can be altered through genetic and environmental perturbations. In a cell-based model of transcriptional activation, increasing or decreasing the cellular concentration of crotonyl-CoA leads to enhanced or diminished gene expression, respectively, which correlates with the levels of histone crotonylation flanking the regulatory elements of activated genes. Our findings support a general principle wherein differential histone acylation (i.e., acetylation versus crotonylation) couples cellular metabolism to the regulation of gene expression.


Asunto(s)
Acilcoenzima A/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Histonas/metabolismo , Macrófagos/inmunología , ARN Mensajero/metabolismo , Activación Transcripcional , Acetato CoA Ligasa/genética , Acetato CoA Ligasa/metabolismo , Acetilación , Acilcoenzima A/genética , Línea Celular , Sistema Libre de Células , Proteína p300 Asociada a E1A/genética , Células HEK293 , Células HeLa , Humanos , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Datos de Secuencia Molecular
19.
Blood ; 136(1): 130-136, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32430495

RESUMEN

Studies of the relationship between the gastrointestinal microbiota and outcomes in allogeneic hematopoietic stem cell transplantation (allo-HCT) have thus far largely focused on early complications, predominantly infection and acute graft-versus-host disease (GVHD). We examined the potential relationship of the microbiome with chronic GVHD (cGVHD) by analyzing stool and plasma samples collected late after allo-HCT using a case-control study design. We found lower circulating concentrations of the microbe-derived short-chain fatty acids (SCFAs) propionate and butyrate in day 100 plasma samples from patients who developed cGVHD, compared with those who remained free of this complication, in the initial case-control cohort of transplant patients and in a further cross-sectional cohort from an independent transplant center. An additional cross-sectional patient cohort from a third transplant center was analyzed; however, serum (rather than plasma) was available, and the differences in SCFAs observed in the plasma samples were not recapitulated. In sum, our findings from the primary case-control cohort and 1 of 2 cross-sectional cohorts explored suggest that the gastrointestinal microbiome may exert immunomodulatory effects in allo-HCT patients at least in part due to control of systemic concentrations of microbe-derived SCFAs.


Asunto(s)
Butiratos/sangre , Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped/microbiología , Propionatos/sangre , Adulto , Aloinjertos , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Estudios de Casos y Controles , Enfermedad Crónica , Disbiosis/etiología , Disbiosis/microbiología , Heces/microbiología , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Metaboloma , Ribotipificación
20.
Nature ; 533(7604): 493-498, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27225120

RESUMEN

Brain metastasis represents a substantial source of morbidity and mortality in various cancers, and is characterized by high resistance to chemotherapy. Here we define the role of the most abundant cell type in the brain, the astrocyte, in promoting brain metastasis. We show that human and mouse breast and lung cancer cells express protocadherin 7 (PCDH7), which promotes the assembly of carcinoma-astrocyte gap junctions composed of connexin 43 (Cx43). Once engaged with the astrocyte gap-junctional network, brain metastatic cancer cells use these channels to transfer the second messenger cGAMP to astrocytes, activating the STING pathway and production of inflammatory cytokines such as interferon-α (IFNα) and tumour necrosis factor (TNF). As paracrine signals, these factors activate the STAT1 and NF-κB pathways in brain metastatic cells, thereby supporting tumour growth and chemoresistance. The orally bioavailable modulators of gap junctions meclofenamate and tonabersat break this paracrine loop, and we provide proof-of-principle that these drugs could be used to treat established brain metastasis.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Uniones Comunicantes/metabolismo , Nucleótidos Cíclicos/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Benzamidas/farmacología , Benzamidas/uso terapéutico , Benzopiranos/farmacología , Benzopiranos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Conexina 43/metabolismo , Resistencia a Antineoplásicos , Femenino , Uniones Comunicantes/efectos de los fármacos , Humanos , Inmunidad Innata , Interferón-alfa/metabolismo , Neoplasias Pulmonares/patología , Ácido Meclofenámico/farmacología , Ácido Meclofenámico/uso terapéutico , Proteínas de la Membrana/metabolismo , Ratones , FN-kappa B/metabolismo , Comunicación Paracrina/efectos de los fármacos , Protocadherinas , Factor de Transcripción STAT1/metabolismo , Factores de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA