Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Genes Dev ; 31(10): 1007-1023, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28611190

RESUMEN

Janus kinase 2 (JAK2) is a central kinase in hematopoietic stem/progenitor cells (HSPCs), and its uncontrolled activation is a prominent oncogenic driver of hematopoietic neoplasms. However, molecular mechanisms underlying the regulation of JAK2 have remained elusive. Here we report that the Casitas B-cell lymphoma (CBL) family E3 ubiquitin ligases down-regulate JAK2 stability and signaling via the adaptor protein LNK/SH2B3. We demonstrated that depletion of CBL/CBL-B or LNK abrogated JAK2 ubiquitination, extended JAK2 half-life, and enhanced JAK2 signaling and cell growth in human cell lines as well as primary murine HSPCs. Built on these findings, we showed that JAK inhibitor (JAKi) significantly reduced aberrant HSPCs and mitigated leukemia development in a mouse model of aggressive myeloid leukemia driven by loss of Cbl and Cbl-b Importantly, primary human CBL mutated (CBLmut ) leukemias exhibited increased JAK2 protein levels and signaling and were hypersensitive to JAKi. Loss-of-function mutations in CBL E3 ubiquitin ligases are found in a wide range of myeloid malignancies, which are diseases without effective treatment options. Hence, our studies reveal a novel signaling axis that regulates JAK2 in normal and malignant HSPCs and suggest new therapeutic strategies for treating CBLmut myeloid malignancies.


Asunto(s)
Janus Quinasa 2/metabolismo , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/fisiopatología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Citocinas/metabolismo , Estabilidad de Enzimas , Células Madre Hematopoyéticas/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Janus Quinasa 2/genética , Leucemia Mieloide Aguda/genética , Proteínas de la Membrana , Ratones , Mutación , Proteolisis , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal/genética , Ubiquitinación
2.
J Immunol ; 197(1): 244-55, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27233963

RESUMEN

Mycobacterium tuberculosis can evade host defense processes, thereby ensuring its survival and pathogenesis. In this study, we investigated the role of nuclear receptor, pregnane X receptor (PXR), in M. tuberculosis infection in human monocyte-derived macrophages. In this study, we demonstrate that PXR augments M. tuberculosis survival inside the host macrophages by promoting the foamy macrophage formation and abrogating phagolysosomal fusion, inflammation, and apoptosis. Additionally, M. tuberculosis cell wall lipids, particularly mycolic acids, crosstalk with human PXR (hPXR) by interacting with its promiscuous ligand binding domain. To confirm our in vitro findings and to avoid the reported species barrier in PXR function, we adopted an in vivo mouse model expressing hPXR, wherein expression of hPXR in mice promotes M. tuberculosis survival. Therefore, pharmacological intervention and designing antagonists to hPXR may prove to be a promising adjunct therapy for tuberculosis.


Asunto(s)
Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Receptores de Esteroides/metabolismo , Tuberculosis/inmunología , Xenobióticos/metabolismo , Animales , Apoptosis , Línea Celular , Supervivencia Celular , Humanos , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fagosomas , Receptor X de Pregnano , Receptores de Esteroides/genética , Transgenes/genética
3.
J Biol Chem ; 290(19): 12222-36, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25809484

RESUMEN

The ability of the active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), to transcriptionally modulate Smads to inhibit Th17 differentiation and experimental autoimmune encephalomyelitis (EAE) has not been adequately studied. This study reports modulation of Smad signaling by the specific binding of the VDR along with its heterodimeric partner RXR to the negative vitamin D response element on the promoter of Smad7, which leads to Smad7 gene repression. The vitamin D receptor-mediated increase in Smad3 expression partially explains the IL10 augmentation seen in Th17 cells. Furthermore, the VDR axis also modulates non-Smad signaling by activating ERK during differentiation of Th17 cells, which inhibits the Th17-specific genes il17a, il17f, il22, and il23r. In vivo EAE experiments revealed that, 1,25(OH)2D3 suppression of EAE correlates with the Smad7 expression in the spleen and lymph nodes. Furthermore, Smad7 expression also correlates well with IL17 and IFNγ expression in CNS infiltered inflammatory T cells. We also observed similar gene repression of Smad7 in in vitro differentiated Th1 cells when cultured in presence of 1,25(OH)2D3. The above canonical and non-canonical pathways in part address the ability of 1,25(OH)2D3-VDR to inhibit EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica , Proteína smad7/antagonistas & inhibidores , Linfocitos T Colaboradores-Inductores/citología , Vitamina D/análogos & derivados , Animales , Células COS , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Chlorocebus aethiops , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Interferón gamma/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Fosforilación , Regiones Promotoras Genéticas , Elementos de Respuesta , Proteína smad7/fisiología , Células Th17 , Transcripción Genética , Vitamina D/química
4.
J Biol Chem ; 290(30): 18304-14, 2015 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25953901

RESUMEN

The orphan nuclear receptor Nr4a2 is known to modulate both inflammatory and metabolic processes, but the mechanism by which it regulates innate inflammatory homeostasis has not been adequately addressed. This study shows that exposure to ligands for Toll-like receptors (TLRs) robustly induces Nr4a2 and that this induction is tightly regulated by the PI3K-Akt signaling axis. Interestingly, exogenous expression of Nr4a2 in macrophages leads to their alternative phenotype with induction of genes that are prototypical M2 markers. Moreover, Nr4a2 transcriptionally activates arginase 1 expression by directly binding to its promoter. Adoptive transfer experiments revealed that increased survival of animals in endotoxin-induced sepsis is Nr4a2-dependent. Thus our data identify a previously unknown role for Nr4a2 in the regulation of macrophage polarization.


Asunto(s)
Inflamación/genética , Macrófagos/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Sepsis/genética , Animales , Polaridad Celular/genética , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/patología , Ligandos , Lipopolisacáridos/toxicidad , Macrófagos/patología , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sepsis/inducido químicamente , Sepsis/metabolismo , Sepsis/patología , Transducción de Señal/genética , Receptores Toll-Like/metabolismo
5.
J Biol Chem ; 290(1): 76-89, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25384979

RESUMEN

GlgB (α-1,4-glucan branching enzyme) is the key enzyme involved in the biosynthesis of α-glucan, which plays a significant role in the virulence and pathogenesis of Mycobacterium tuberculosis. Because α-glucans are implicated in the survival of both replicating and non-replicating bacteria, there exists an exigent need for the identification and development of novel inhibitors for targeting enzymes, such as GlgB, involved in this pathway. We have used the existing structural information of M. tuberculosis GlgB for high throughput virtual screening and molecular docking. A diverse database of 330,000 molecules was used for identifying novel and efficacious therapeutic agents for targeting GlgB. We also used three-dimensional shape as well as two-dimensional similarity matrix methods to identify diverse molecular scaffolds that inhibit M. tuberculosis GlgB activity. Virtual hits were generated after structure and ligand-based screening followed by filters based on interaction with human GlgB and in silico pharmacokinetic parameters. These hits were experimentally evaluated and resulted in the discovery of a number of structurally diverse chemical scaffolds that target M. tuberculosis GlgB. Although a number of inhibitors demonstrated in vitro enzyme inhibition, two compounds in particular showed excellent inhibition of in vivo M. tuberculosis survival and its ability to get phagocytosed. This work shows that in silico docking and three-dimensional chemical similarity could be an important therapeutic approach for developing inhibitors to specifically target the M. tuberculosis GlgB enzyme.


Asunto(s)
Enzima Ramificadora de 1,4-alfa-Glucano/antagonistas & inhibidores , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Descubrimiento de Drogas , Mycobacterium tuberculosis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Enzima Ramificadora de 1,4-alfa-Glucano/química , Enzima Ramificadora de 1,4-alfa-Glucano/genética , Enzima Ramificadora de 1,4-alfa-Glucano/metabolismo , Antituberculosos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Bases de Datos Farmacéuticas , Bases de Datos de Proteínas , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glucanos/química , Glucanos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/crecimiento & desarrollo , Fagocitosis/efectos de los fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Homología Estructural de Proteína , Relación Estructura-Actividad , Interfaz Usuario-Computador
6.
J Biol Chem ; 288(15): 10692-702, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23449984

RESUMEN

Nuclear receptors modulate macrophage effector functions, which are imperative for clearance or survival of mycobacterial infection. The adopted orphan nuclear receptor Rev-erbα is a constitutive transcriptional repressor as it lacks AF2 domain and was earlier shown to be present in macrophages. In the present study, we highlight the differences in the relative subcellular localization of Rev-erbα in monocytes and macrophages. The nuclear localization of Rev-erbα in macrophages is subsequent to monocyte differentiation. Expression analysis of Rev-erbα elucidated it to be considerably more expressed in M1 phenotype in comparison with M2. Rev-erbα overexpression augments antimycobacterial properties of macrophage by keeping IL10 in a basal repressed state. Further, promoter analysis revealed that IL10 promoter harbors a Rev-erbα binding site exclusive to humans and higher order primates and not mouse, demonstrating a species barrier in its functionality. This direct gene repression is mediated by recruitment of co-repressors NCoR and HDAC3. In addition, our data elucidate that its overexpression reduced the survival of intracellular pathogen Mycobacterium tuberculosis by enhancing phagosome lysosome maturation, an event resulting from IL10 repression. Thus, these findings suggest that Rev-erbα bestows protection against mycobacterial infection by direct gene repression of IL10 and thus provide a novel target in modulating macrophage microbicidal properties.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-10/biosíntesis , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Elementos de Respuesta , Animales , Línea Celular , Femenino , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Mycobacterium tuberculosis/inmunología , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/inmunología , Especificidad de la Especie , Tuberculosis/genética , Tuberculosis/inmunología , Tuberculosis/metabolismo , Tuberculosis/patología
7.
J Immunol ; 188(11): 5593-603, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22544925

RESUMEN

Mycobacterium tuberculosis-macrophage interactions are key to pathogenesis and clearance of these bacteria. Although interactions between M. tuberculosis-associated lipids and TLRs, non-TLRs, and opsonic receptors have been investigated, interactions of these lipids and infected macrophage lipid repertoire with lipid-sensing nuclear receptors expressed in macrophages have not been addressed. In this study, we report that M. tuberculosis-macrophage lipids can interact with host peroxisome proliferator-activated receptor γ and testicular receptor 4 to ensure survival of the pathogen by modulating macrophage function. These two lipid-sensing nuclear receptors create a foamy niche within macrophage by modulating oxidized low-density lipoprotein receptor CD36, phagolysosomal maturation block by induction of IL-10, and a blunted innate response by alternative polarization of the macrophages, which leads to survival of M. tuberculosis. These results also suggest possible heterologous ligands for peroxisome proliferator-activated receptor γ and testicular receptor 4 and are suggestive of adaptive or coevolution of the host and pathogen. Relative mRNA expression levels of these receptors in PBMCs derived from clinical samples convincingly implicate them in tuberculosis susceptibility. These observations expose a novel paradigm in the pathogenesis of M. tuberculosis amenable for pharmacological modulation.


Asunto(s)
Células Espumosas/inmunología , Células Espumosas/microbiología , Metabolismo de los Lípidos/inmunología , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/inmunología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , PPAR gamma/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Células Espumosas/metabolismo , Humanos , Ligandos
8.
J Infect Dis ; 206(3): 366-76, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22615313

RESUMEN

BACKGROUND: Mycobacterium tuberculosis, the causative agent of tuberculosis, has a remarkable ability to usurp its host's innate immune response, killing millions of infected people annually. One approach to manage infection is prevention through the use of natural agents. In this regard, stem bromelain (SBM), a pharmacologically active member of the sulfhydryl proteolytic enzyme family, obtained from Ananas comosus and possessing a remarkable ability to induce the innate and acquired immune systems, is important. METHODS: We evaluated SBM's ability to induce apoptosis and free-radical generation in macrophages. We also studied antimycobacterial properties of SBM and its effect on foamy macrophages. RESULTS: SBM treatment of peritoneal macrophages resulted in the upregulation of proapoptotic proteins and downregulation of antiapoptotic proteins. Additionally, SBM treatment activated macrophages, curtailed the levels of free glutathione, and augmented the production of hydrogen peroxide, superoxide anion, peroxynitrite, and nitric oxide. SBM cleaves CD36 and reduced the formation of foam cells, the hallmark of M. tuberculosis infection. These conditions created an environment for the increased clearance of M. tuberculosis. CONCLUSIONS: Together these data provide a mechanism for antimycobacterial activity of SBM and provide important insights for the use of cysteine proteases as immunomodulatory agents.


Asunto(s)
Antituberculosos/farmacología , Apoptosis/efectos de los fármacos , Bromelaínas/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Secuencia de Aminoácidos , Ananas/química , Animales , Bromelaínas/química , Antígenos CD36/química , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Anotación de Secuencia Molecular , Especies Reactivas de Oxígeno/metabolismo
9.
Mol Ther Nucleic Acids ; 31: 452-465, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36852088

RESUMEN

Transcriptional enhancers can be in physical proximity of their target genes via chromatin looping. The enhancer at the ß-globin locus (locus control region [LCR]) contacts the fetal-type (HBG) and adult-type (HBB) ß-globin genes during corresponding developmental stages. We have demonstrated previously that forcing proximity between the LCR and HBG genes in cultured adult-stage erythroid cells can activate HBG transcription. Activation of HBG expression in erythroid cells is of benefit to patients with sickle cell disease. Here, using the ß-globin locus as a model, we provide proof of concept at the organismal level that forced enhancer rewiring might present a strategy to alter gene expression for therapeutic purposes. Hematopoietic stem and progenitor cells (HSPCs) from mice bearing human ß-globin genes were transduced with lentiviral vectors expressing a synthetic transcription factor (ZF-Ldb1) that fosters LCR-HBG contacts. When engrafted into host animals, HSPCs gave rise to adult-type erythroid cells with elevated HBG expression. Vectors containing ZF-Ldb1 were optimized for activity in cultured human and rhesus macaque erythroid cells. Upon transplantation into rhesus macaques, erythroid cells from HSPCs expressing ZF-Ldb1 displayed elevated HBG production. These findings in two animal models suggest that forced redirection of gene-regulatory elements may be used to alter gene expression to treat disease.

10.
Blood Adv ; 6(3): 731-745, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34844262

RESUMEN

Hematopoietic stem cell transplantation (HSCT) remains the only curative treatment for a variety of hematological diseases. Allogenic HSCT requires hematopoietic stem cells (HSCs) from matched donors and comes with cytotoxicity and mortality. Recent advances in genome modification of HSCs have demonstrated the possibility of using autologous HSCT-based gene therapy to alleviate hematologic symptoms in monogenic diseases, such as the inherited bone marrow failure (BMF) syndrome Fanconi anemia (FA). However, for FA and other BMF syndromes, insufficient HSC numbers with functional defects results in delayed hematopoietic recovery and increased risk of graft failure. We and others previously identified the adaptor protein LNK (SH2B3) as a critical negative regulator of murine HSC homeostasis. However, whether LNK controls human HSCs has not been studied. Here, we demonstrate that depletion of LNK via lentiviral expression of miR30-based short hairpin RNAs results in robust expansion of transplantable human HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Importantly, LNK depletion enhances cytokine-mediated JAK/STAT activation in CD34+ hematopoietic stem and progenitor cells (HSPCs). Moreover, we demonstrate that LNK depletion expands primary HSPCs associated with FA. In xenotransplant, engraftment of FANCD2-depleted FA-like HSCs was markedly improved by LNK inhibition. Finally, targeting LNK in primary bone marrow HSPCs from FA patients enhanced their colony forming potential in vitro. Together, these results demonstrate the potential of targeting LNK to expand HSCs to improve HSCT and HSCT-based gene therapy.


Asunto(s)
Anemia de Fanconi , Trasplante de Células Madre Hematopoyéticas , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD34/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones
11.
Autophagy ; 17(9): 2273-2289, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917126

RESUMEN

Macrophage derived foam cells in atherosclerotic plaques are the major factor responsible for the pathogenesis of atherosclerosis (AS). During advanced AS, macrophage-specific macroautophagy/autophagy is dysfunctional. 1, 25-dihydroxy vitamin D3 (VitD3) and its receptor VDR (vitamin D receptor) are reported to inhibit foam cell formation and induce autophagy; however, the role of VitD3-VDR-induced autophagy and foam cell formation in AS has not been explored. Here we find that VitD3 significantly recovered oxidized low-density lipoprotein-impaired autophagy, as well as increased autophagy-mediated lipid breakdown in mouse bone marrow-derived macrophages and human monocyte-derived macrophages, thus inhibiting the conversion of macrophages into foam cells. Importantly, VitD3 functions through its receptor VDR to upregulate autophagy and attenuate the accumulation of lipids in macrophages. Moreover, this study is the first occasion to report the interesting link between VitD3 signaling and PTPN6/SHP-1 (protein tyrosine phosphatase non-receptor type 6) in macrophages. VitD3-induced autophagy was abrogated in the presence of the PTPN6/Ptpn6 shRNA or inhibitor. VDR along with RXRA (retinoid X receptor alpha), and NCOA1 (nuclear receptor coactivator 1), are recruited to a specific response element located on the gene promoter and induce PTPN6 expression. PTPN6 contributes to VitD3-mediated autophagy by regulating autophagy-related genes via activation of MAPK1 (mitogen-activated protein kinase 1) and CEBPB (CCAAT enhancer binding protein beta). Furthermore, expression of PTPN6 is also crucial for VitD3-mediated inhibition of macrophage foam cell formation through autophagy. Thus, VitD3-VDR-PTPN6 axis-regulated autophagy attenuates foam cell formation in macrophages.


Asunto(s)
Autofagia , Colecalciferol , Células Espumosas , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Receptores de Calcitriol , Animales , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de Calcitriol/metabolismo
12.
Arch Biochem Biophys ; 499(1-2): 26-31, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20494823

RESUMEN

Stem bromelain (SBM), a therapeutic protein, is rapidly absorbed across the gut epithelium. Because SBM encounters an alkaline pH at its principal site of absorption, we investigated the alkaline-induced denaturation of SBM. From pH 7 to 10, the protein's secondary structure remained the same, although a slight loss of tertiary structure was observed. Above pH 10, there was a significant and irreversible loss of secondary and tertiary structure. At pH 10, SBM showed enhanced tryptophan fluorescence, however, the number of accessible tryptophans remained the same. The thermodynamics of temperature transition at pH 7 and 10 were strikingly different, with the former showing a two-phase transition endotherm, and the latter a broad non-two-state transition. At pH 10, SBM showed a significant increase in 8-anilino-1-naphthalene-sulfonate binding relative to the native state, suggestive of a specific molten globule (SMG) state. These studies suggest a distinct conformational rearrangement in SBM, at the protein's isoelectric point.


Asunto(s)
Bromelaínas/química , Proteínas de Plantas/química , Ananas/enzimología , Bromelaínas/farmacocinética , Dicroismo Circular , Guanidina , Calor , Humanos , Concentración de Iones de Hidrógeno , Técnicas In Vitro , Absorción Intestinal , Punto Isoeléctrico , Fitoterapia , Proteínas de Plantas/farmacocinética , Conformación Proteica , Desnaturalización Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia , Triptófano/química
13.
Nat Cell Biol ; 22(10): 1170-1179, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32989246

RESUMEN

SIRT1 (Sir2) is an NAD+-dependent deacetylase that plays critical roles in a broad range of biological events, including metabolism, the immune response and ageing1-5. Although there is strong interest in stimulating SIRT1 catalytic activity, the homeostasis of SIRT1 at the protein level is poorly understood. Here we report that macroautophagy (hereafter referred to as autophagy), a catabolic membrane trafficking pathway that degrades cellular components through autophagosomes and lysosomes, mediates the downregulation of mammalian SIRT1 protein during senescence and in vivo ageing. In senescence, nuclear SIRT1 is recognized as an autophagy substrate and is subjected to cytoplasmic autophagosome-lysosome degradation, via the autophagy protein LC3. Importantly, the autophagy-lysosome pathway contributes to the loss of SIRT1 during ageing of several tissues related to the immune and haematopoietic system in mice, including the spleen, thymus, and haematopoietic stem and progenitor cells, as well as in CD8+CD28- T cells from aged human donors. Our study reveals a mechanism in the regulation of the protein homeostasis of SIRT1 and suggests a potential strategy to stabilize SIRT1 to promote productive ageing.


Asunto(s)
Autofagosomas/metabolismo , Autofagia , Senescencia Celular , Proteínas Asociadas a Microtúbulos/metabolismo , Sirtuina 1/antagonistas & inhibidores , Células Madre/citología , Linfocitos T/patología , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Femenino , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/genética , Persona de Mediana Edad , Sirtuina 1/genética , Sirtuina 1/metabolismo , Células Madre/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
14.
Nat Commun ; 9(1): 3915, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254368

RESUMEN

Fanconi anemia (FA) is a bone marrow failure (BMF) syndrome that arises from mutations in a network of FA genes essential for DNA interstrand crosslink (ICL) repair and replication stress tolerance. While allogeneic stem cell transplantation can replace defective HSCs, interventions to mitigate HSC defects in FA do not exist. Remarkably, we reveal here that Lnk (Sh2b3) deficiency restores HSC function in Fancd2-/- mice. Lnk deficiency does not impact ICL repair, but instead stabilizes stalled replication forks in a manner, in part, dependent upon alleviating blocks to cytokine-mediated JAK2 signaling. Lnk deficiency restores proliferation and survival of Fancd2-/- HSCs, while reducing replication stress and genomic instability. Furthermore, deletion of LNK in human FA-like HSCs promotes clonogenic growth. These findings highlight a new role for cytokine/JAK signaling in promoting replication fork stability, illuminate replication stress as a major underlying origin of BMF in FA, and have strong therapeutic implications.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Trasplante de Médula Ósea , Proliferación Celular/genética , Células Cultivadas , Reparación del ADN/genética , Replicación del ADN/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Proteínas de la Membrana , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados
15.
Biol Rev Camb Philos Soc ; 91(2): 429-51, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-25651938

RESUMEN

Autophagy is a process that maintains the equilibrium between biosynthesis and the recycling of cellular constituents; it is critical for avoiding the pathophysiology that results from imbalance in cellular homeostasis. Recent reports indicate the need for the design of high-throughput screening assays to identify targets and small molecules for autophagy modulation. For such screening, however, a better understanding of the regulation of autophagy is essential. In addition to regulation by various signalling cascades, regulation of gene expression by transcription factors is also critical. This review focuses on the various transcription factors as well as the corresponding signalling molecules that act together to translate the stimuli to effector molecules that up- or downregulate autophagy. This review rationalizes the importance of these transcription factors functioning in tandem with cognate signalling molecules and their interfaces as possible therapeutic targets for more specific pharmacological interventions.


Asunto(s)
Autofagia/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Animales , Autofagia/efectos de los fármacos , Regulación de la Expresión Génica/fisiología
16.
Autophagy ; 11(11): 1987-1997, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26390081

RESUMEN

NR1D1 (nuclear receptor subfamily 1, group D, member 1), an adopted orphan nuclear receptor, is widely known to orchestrate the expression of genes involved in various biological processes such as adipogenesis, skeletal muscle differentiation, and lipid and glucose metabolism. Emerging evidence suggests that various members of the nuclear receptor superfamily perform a decisive role in the modulation of autophagy. Recently, NR1D1 has been implicated in augmenting the antimycobacterial properties of macrophages and providing protection against Mycobacterium tuberculosis infection by downregulating the expression of the IL10 gene in human macrophages. This antiinfective property of NR1D1 suggests the need for an improved understanding of its role in other host-associated antimycobacterial pathways. The results presented here demonstrate that in human macrophages either ectopic expression of NR1D1 or treatment with its agonist, GSK4112, enhanced the number of acidic vacuoles as well as the level of MAP1LC3-II, a signature molecule for determination of autophagy progression, in a concentration- and time-dependent manner. Conversely, a decrease in NR1D1 in knockdown cells resulted in the reduced expression of lysosomal-associated membrane protein 1, LAMP1, commensurate with a decrease in the level of transcription factor EB, TFEB. This is indicative of that NR1D1 may have a regulatory role in lysosome biogenesis. NR1D1 being a repressor, its positive regulation on LAMP1 and TFEB is suggestive of an indirect byzantine mechanism of action. Its role in the modulation of autophagy and lysosome biogenesis together with its ability to repress IL10 gene expression supports the theory that NR1D1 has a pivotal antimycobacterial function in human macrophages.

17.
Int J Biol Macromol ; 77: 168-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25799883

RESUMEN

Protein methyltransferase (CheR) catalyzes the methylation of the cytosolic domain of the membrane bound chemotaxis receptors, and plays a pivotal role in the chemotactic signal transduction pathway in bacteria. Crystal structure of CheR is available only from the gram-negative bacterium Salmonella typhimurium (StCheR), which contain a catalytic C-terminal domain, encompassing a ß-subdomain, connected via a linker to the N-terminal domain. The structural-functional similitude between CheR of the gram-negative and the gram-positive bacteria remains obscure. We investigated CheR, from a gram-positive bacterium, Bacillus subtilis (BsCheR), and have identified the functional roles of its N-terminal domain, by using the in silico molecular modeling and docking approach along with mass spectrophotometry and sequence analysis. The structural studies established that the N-terminal domain directly bound to S-Adenosyl-l-homocysteine (SAH). Structural and sequence analyses revealed that the α2 helix of the N-terminal domain was involved in the recognition of the methylation site of the chemotactic receptor. Additionally, immunoblot analysis showed that the purified BsCheR was phosphorylated. Further, mass spectrometry studies detected the phosphorylation at Thr3 position in the N-terminal domain of BsCheR. Phosphorylation of BsCheR suggested a regulatory role of the N-terminal domain, analogous to its antagonistic enzyme, the chemotaxis-specific methylesterase (CheB).


Asunto(s)
Bacillus subtilis/enzimología , Proteína Metiltransferasas/metabolismo , Proteómica , Secuencia de Aminoácidos , Bacillus subtilis/genética , Dominio Catalítico , Simulación por Computador , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Proteína Metiltransferasas/química , Proteína Metiltransferasas/genética , Estructura Secundaria de Proteína , S-Adenosilhomocisteína/metabolismo
18.
Int J Biol Macromol ; 49(4): 536-42, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21736896

RESUMEN

Stem bromelain (SBM) is a therapeutic protein that has been studied for alkaline denaturation in the intestines, the principal site of its absorption. In this study, we investigated fluorinated alcohol 2,2,2-trifluoroethanol (TFE)-induced conformational changes in the specific/pre-molten globule (SMG) state of SBM observed at pH 10 by spectroscopic methods. Far-UV circular dichroism (CD) spectra showed that the protein retained its native-like secondary structure at TFE concentrations of up to 30% with a pronounced minimum at 222 nm, characteristic of a helix. However, addition of slightly higher TFE concentrations (≥40%) resulted in an ∼2.5-fold induction of this helical feature and a time-dependent increase in non-amyloidic turbidity as evidenced by turbidometric, Congo red-binding, and Thioflavin T (ThT)-binding studies. Near-UV CD spectra suggested a gradual but significant loss of tertiary structure at 10-30% TFE. Tryptophan studies showed blue-shifted fluorescence, although the number of accessible tryptophans remained the same up to 30% TFE. The SMG showed enhanced binding of the fluorescent probe 1-anilino-8-naphthalene sulfonic acid (ANS) up to 30% TFE, beyond which binding plateaued. Thermal and guanidine hydrochloride (GdnHCl) transition studies in the near-UV range indicated a single cooperative transition for the SMG state in the presence of 30% TFE, similar to that observed for native SBM at pH 7.0 (although with different T(m)s), unlike the SMG state. TFE (30%) appeared to induce native-like stability to the original SMG. These observations suggest a transformation of the SMG to a characteristic molten globule (MG) conformation at 30% TFE, possibly due to TFE-induced rearrangement of hydrophobic interactions at the protein's isoelectric point.


Asunto(s)
Amiloide/química , Bromelaínas/química , Bromelaínas/metabolismo , Trifluoroetanol/farmacología , Absorción/efectos de los fármacos , Acrilamida/química , Naftalenosulfonatos de Anilina/metabolismo , Dicroismo Circular , Estabilidad de Enzimas/efectos de los fármacos , Guanidina/farmacología , Punto Isoeléctrico , Nefelometría y Turbidimetría , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Desnaturalización Proteica/efectos de los fármacos , Espectrometría de Fluorescencia , Temperatura , Triptófano/metabolismo
19.
Int J Biochem Cell Biol ; 42(6): 938-47, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20156582

RESUMEN

Stem bromelain is a proteolytic phytoprotein with a variety of therapeutic effects. Understanding its structural properties could provide insight into the mechanisms underlying its clinical utility. Stem bromelain was evaluated for its conformational and folding properties at the pH conditions it encounters when administered orally. It exists as a partially folded intermediate at pH 2.0. The conformational changes to this intermediate state were evaluated using fluorinated alcohols known to induce changes similar to those seen in vivo. Studies using circular dichroism, fluorescence emission spectroscopy, binding of the hydrophobic dye 1-anilino-8-naphthalene sulfonic acid and mass spectrometry indicate that treatment with 10-30% hexafluoroisopropanol induces the partially folded intermediate to adopt much of the native protein's secondary structure, but only a rudimentary tertiary structure, characteristic of the molten globule state. Addition of slightly higher concentrations of hexafluoroisopropanol caused transformation from an alpha-helix to a beta-sheet and induced formation of a compact nonnative structure. This nonnative form was more inhibitory of cell survival than either the native or the partially folded intermediate forms, as measured by enhanced suppression of proliferative cues (e.g., extracellular-signal-regulated kinase) and initiation of apoptotic events. The nonnative form also showed better antitumorigenic properties, as evaluated using an induced two-stage mouse skin papilloma model. In contrast, the nonnative state showed only a fraction of the proteolytic activity of the native form. This study demonstrates that hexafluoroisopropanol can induce a conformational change in stem bromelain to a form with potentially useful therapeutic properties different from those of the native protein.


Asunto(s)
Antineoplásicos/farmacología , Bromelaínas/farmacología , Fibroblastos/efectos de los fármacos , Papiloma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Células 3T3 , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Bromelaínas/química , Proliferación Celular/efectos de los fármacos , Dicroismo Circular , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Papiloma/patología , Propanoles/química , Estructura Secundaria de Proteína , Relación Estructura-Actividad Cuantitativa , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA