Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36214220

RESUMEN

Intercellular communication is critical for homeostasis in mammalian systems, including the gastrointestinal (GI) tract. Exosomes are nanoscale lipid extracellular vesicles that mediate communication between many cell types. Notably, the roles of immune cell exosomes in regulating GI homeostasis and inflammation are largely uncharacterized. By generating mouse strains deficient in cell-specific exosome production, we demonstrate deletion of the small GTPase Rab27A in CD11c+ cells exacerbated murine colitis, which was reversible through administration of DC-derived exosomes. Profiling RNAs within colon exosomes revealed a distinct subset of miRNAs carried by colon- and DC-derived exosomes. Among antiinflammatory exosomal miRNAs, miR-146a was transferred from gut immune cells to myeloid and T cells through a Rab27-dependent mechanism, targeting Traf6, IRAK-1, and NLRP3 in macrophages. Further, we have identified a potentially novel mode of exosome-mediated DC and macrophage crosstalk that is capable of skewing gut macrophages toward an antiinflammatory phenotype. Assessing clinical samples, RAB27A, select miRNAs, and RNA-binding proteins that load exosomal miRNAs were dysregulated in ulcerative colitis patient samples, consistent with our preclinical mouse model findings. Together, our work reveals an exosome-mediated regulatory mechanism underlying gut inflammation and paves the way for potential use of miRNA-containing exosomes as a novel therapeutic for inflammatory bowel disease.


Asunto(s)
Antígenos CD11 , Colitis , Exosomas , Inflamación , Células Mieloides , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Colitis/genética , Colitis/inmunología , Exosomas/genética , Exosomas/inmunología , Inflamación/genética , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Lípidos , Mamíferos/genética , Mamíferos/inmunología , Ratones , MicroARNs/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Células Mieloides/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Factor 6 Asociado a Receptor de TNF/inmunología
2.
Biochem Soc Trans ; 49(5): 2021-2035, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623375

RESUMEN

The RAS superfamily of small GTPases regulates major physiological cellular processes. Mutation or deregulation of these small GTPases, their regulators and/or their effectors are associated with many diseases including cancer. Hence, targeting these classes of proteins is an important therapeutic strategy in cancer. This has been recently achieved with the approval of the first KRASG12C covalent inhibitors for the clinic. However, many other mutants and small GTPases are still considered as 'undruggable' with small molecule inhibitors because of a lack of well-defined pocket(s) at their surface. Therefore, alternative therapeutic strategies have been developed to target these proteins. In this review, we discuss the use of intracellular antibodies and derivatives - reagents that bind their antigen inside the cells - for the discovery of novel inhibitory mechanisms, targetable features and therapeutic strategies to inhibit small GTPases and their downstream pathways. These reagents are also versatile tools used to better understand the biological mechanisms regulated by small GTPases and to accelerate the drug discovery process.


Asunto(s)
Anticuerpos/metabolismo , Proteínas de Repetición de Anquirina Diseñadas/metabolismo , Descubrimiento de Drogas/métodos , Proteínas de Unión al GTP Monoméricas/metabolismo , Neoplasias/enzimología , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas de Repetición de Anquirina Diseñadas/farmacología , Humanos , Terapia Molecular Dirigida/métodos , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/inmunología , Neoplasias/tratamiento farmacológico , Unión Proteica , Proteolisis/efectos de los fármacos , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología
3.
Arthritis Rheumatol ; 73(7): 1200-1210, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33452873

RESUMEN

OBJECTIVE: ZAP-70W163C BALB/c (SKG) mice develop reactive arthritis (ReA) following infection with Chlamydia muridarum. Since intracellular pathogens enhance their replicative fitness in stressed host cells, we examined how myeloid cells infected with C muridarum drive arthritis. METHODS: SKG, Il17a-deficient SKG, and BALB/c female mice were infected with C muridarum or C muridarum luciferase in the genitals. C muridarum dissemination was assessed by in vivo imaging or genomic DNA amplification. Macrophages were depleted using clodronate liposomes. Anti-tumor necrosis factor (anti-TNF) and anti-interleukin-23p19 (anti-IL-23p19) were administered after infection or arthritis onset. Gene expression of Hspa5, Tgtp1, Il23a, Il17a, Il12b, and Tnf was compared in SKG mice and BALB/c mice. RESULTS: One week following infection with C muridarum, macrophages and neutrophils were observed to have infiltrated the uteri of mice and were also shown to have carried C muridarum DNA to the spleen. C muridarum load was higher in SKG mice than in BALB/c mice. Macrophage depletion was shown to reduce C muridarum load and prevent development of arthritis. Compared with BALB/c mice, expression of Il23a and Il17a was increased in the uterine and splenic neutrophils of SKG mice. The presence of anti-IL-23p19 during infection or Il17a deficiency suppressed arthritis. Tnf was overexpressed in the joints of SKG mice within 1 week postinfection, and persisted beyond the first week. TNF inhibition during infection or at arthritis onset suppressed the development of arthritis. Levels of endoplasmic reticulum stress were constitutively increased in the joints of SKG mice but were induced, in conjunction with immunity-related GTPase, by C muridarum infection in the uterus. CONCLUSION: C muridarum load is higher in SKG mice than in BALB/c mice. Whereas proinflammatory IL-23 produced by neutrophils contributes to the initiation of C muridarum-mediated ReA, macrophage depletion reduces C muridarum dissemination to other tissues, tissue burden, and the development of arthritis. TNF inhibition was also shown to suppress arthritis development. Our data suggest that enhanced bacterial dissemination in macrophages of SKG mice drives the TNF production needed for persistent arthritis.


Asunto(s)
Artritis Reactiva/inmunología , Infecciones por Chlamydia/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Interleucina-23/inmunología , Macrófagos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Artritis Experimental/genética , Artritis Reactiva/genética , Chlamydia muridarum , Chaperón BiP del Retículo Endoplásmico , Femenino , Perfilación de la Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Subunidad p19 de la Interleucina-23/genética , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Factor de Necrosis Tumoral alfa/genética , Proteína Tirosina Quinasa ZAP-70/genética
4.
J Exp Med ; 217(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31649036

RESUMEN

Foxp3+ regulatory T (T reg) cells are pivotal regulators of immune tolerance, with T cell receptor (TCR)-driven activated T reg (aT reg) cells playing a central role; yet how TCR signaling propagates to control aT reg cell responses remains poorly understood. Here we show that TCR signaling induces expression of amino acid transporters, and renders amino acid-induced activation of mTORC1 in aT reg cells. T reg cell-specific ablation of the Rag family small GTPases RagA and RagB impairs amino acid-induced mTORC1 signaling, causing defective amino acid anabolism, reduced T reg cell proliferation, and a rampant autoimmune disorder similar in severity to that triggered by T reg cell-specific TCR deficiency. Notably, T reg cells in peripheral tissues, including tumors, are more sensitive to Rag GTPase-dependent nutrient sensing. Ablation of RagA alone impairs T reg cell accumulation in the tumor, resulting in enhanced antitumor immunity. Thus, nutrient mTORC1 signaling is an essential component of TCR-initiated T reg cell reprogramming, and Rag GTPase activities may be titrated to break tumor immune tolerance.


Asunto(s)
Tolerancia Inmunológica/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Nutrientes/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Sistemas de Transporte de Aminoácidos/inmunología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al GTP Monoméricas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/inmunología
5.
J Exp Med ; 216(10): 2427-2447, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31324740

RESUMEN

Signal strength controls the outcome of αß T cell selection in the thymus, resulting in death if the affinity of the rearranged TCR is below the threshold for positive selection, or if the affinity of the TCR is above the threshold for negative selection. Here we show that deletion of the GTPase RRAS2 results in exacerbated negative selection and above-normal expression of positive selection markers. Furthermore, Rras2-/- mice are resistant to autoimmunity both in a model of inflammatory bowel disease (IBD) and in a model of myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). We show that MOG-specific T cells in Rras2-/- mice have reduced affinity for MOG/I-Ab tetramers, suggesting that enhanced negative selection leads to selection of TCRs with lower affinity for the self-MOG peptide. An analysis of the TCR repertoire shows alterations that mostly affect the TCRα variable (TRAV) locus with specific VJ combinations and CDR3α sequences that are absent in Rras2-/- mice, suggesting their involvement in autoimmunity.


Asunto(s)
Selección Clonal Mediada por Antígenos , Encefalomielitis Autoinmune Experimental/inmunología , Reordenamiento Génico de la Cadena alfa de los Receptores de Antígenos de los Linfocitos T/inmunología , Proteínas de la Membrana/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Animales , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/inmunología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/genética , Glicoproteína Mielina-Oligodendrócito/efectos adversos , Glicoproteína Mielina-Oligodendrócito/farmacología
6.
PLoS Pathog ; 12(8): e1005829, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27541004

RESUMEN

Macrophages are a heterogeneous cell population strongly influenced by differentiation stimuli that become susceptible to HIV-1 infection after inactivation of the restriction factor SAMHD1 by cyclin-dependent kinases (CDK). Here, we have used primary human monocyte-derived macrophages differentiated through different stimuli to evaluate macrophage heterogeneity on cell activation and proliferation and susceptibility to HIV-1 infection. Stimulation of monocytes with GM-CSF induces a non-proliferating macrophage population highly restrictive to HIV-1 infection, characterized by the upregulation of the G1/S-specific cyclin D2, known to control early steps of cell cycle progression. Knockdown of cyclin D2, enhances HIV-1 replication in GM-CSF macrophages through inactivation of SAMHD1 restriction factor by phosphorylation. Co-immunoprecipitation experiments show that cyclin D2 forms a complex with CDK4 and p21, a factor known to restrict HIV-1 replication by affecting the function of the downstream cascade that leads to SAMHD1 deactivation. Thus, we demonstrate that cyclin D2 acts as regulator of cell cycle proteins affecting SAMHD1-mediated HIV-1 restriction in non-proliferating macrophages.


Asunto(s)
Ciclina D2/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Macrófagos/inmunología , Animales , Proliferación Celular , Quinasa 4 Dependiente de la Ciclina/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Macrófagos/virología , Ratones , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
7.
Sci Rep ; 6: 22629, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26936683

RESUMEN

Restriction of HIV-1 in myeloid-lineage cells is attributed in part to the nucleotidase activity of the SAM-domain and HD-domain containing protein (SAMHD1), which depletes free nucleotides, blocking reverse transcription. In the same cells, the Vpx protein of HIV-2 and most SIVs counteracts SAMHD1. Both Type I and II interferons may stimulate SAMHD1 transcription. The contributions of SAMHD1 to retroviral restriction in the central nervous system (CNS) have been the subject of limited study. We hypothesized that SAMHD1 would respond to interferon in the SIV-infected CNS but would not control virus due to SIV Vpx. Accordingly, we investigated SAMHD1 transcript abundance and association with the Type I interferon response in an SIV model. SAMHD1 transcript levels were IFN responsive, increasing during acute phase infection and decreasing during a more quiescent phase, but generally remaining elevated at all post-infection time points. In vitro, SAMHD1 transcript was abundant in macaque astrocytes and further induced by Type I interferon, while IFN produced a weaker response in the more permissive environment of the macrophage. We cannot rule out a contribution of SAMHD1 to retroviral restriction in relatively non-permissive CNS cell types. We encourage additional research in this area, particularly in the context of HIV-1 infection.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/metabolismo , Macrófagos/metabolismo , Proteínas de Unión al GTP Monoméricas/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Regulación hacia Arriba/inmunología , Carga Viral/inmunología , Animales , Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/virología , Femenino , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Macaca nemestrina , Macrófagos/inmunología , Macrófagos/virología , Masculino , Proteínas de Unión al GTP Monoméricas/biosíntesis , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Virus de la Inmunodeficiencia de los Simios/inmunología
8.
Biol Chem ; 397(4): 373-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26733158

RESUMEN

Sterile alpha motif and histidine-aspartate domain containing protein 1 (SAMHD1) is a triphosphohydrolase that catalyzes the conversion of deoxyribonucleoside triphosphate to deoxyribonucleoside and triphosphate. SAMHD1 has been a recent focus of study since it was identified as a potent human immunodeficiency virus-1 (HIV-1) restriction factor in the intrinsic antiviral immune system. Recent biochemical and biological studies have suggested that SAMHD1 restricts HIV-1 infection in non-cycling cells by limiting the pool of deoxyribonucleoside triphosphates, thereby interfering with HIV-1 reverse transcription. SAMHD1 also possesses single-stranded DNA and RNA binding activity, with reported nuclease activity, conferring additional HIV-1 restriction function. This review summarizes current knowledge regarding the structure of SAMHD1 and the regulation of its function in HIV-1 restriction.


Asunto(s)
Infecciones por VIH/virología , VIH-1/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/inmunología , Humanos , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
9.
PLoS One ; 10(6): e0131139, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26091527

RESUMEN

BACKGROUND: Several soluble factors have been reported to have the capacity of inhibiting HIV replication at different steps of the virus life cycle, without eliminating infected cells and through enhancement of specific cellular mechanisms. Yet, it is unclear if these antiviral factors play a role in the protection from HIV infection or in the control of viral replication. Here we evaluated two cohorts: i) one of 58 HIV-exposed seronegative individuals (HESNs) who were compared with 59 healthy controls (HCs), and ii) another of 13 HIV-controllers who were compared with 20 HIV-progressors. Peripheral blood, oral and genital mucosa and gut-associated lymphoid tissue (GALT) samples were obtained to analyze the mRNA expression of ELAFIN, APOBEC3G, SAMHD1, TRIM5α, RNase 7 and SerpinA1 using real-time PCR. RESULTS: HESNs exhibited higher expression of all antiviral factors in peripheral blood mononuclear cells (PBMCs), oral or genital mucosa when compared with HCs. Furthermore, HIV-controllers exhibited higher levels of SerpinA1 in GALT. CONCLUSIONS: These findings suggest that the activity of these factors is compartmentalized and that these proteins have a predominant role depending on the tissue to avoid the infection, reduce the viral load and modulate the susceptibility to HIV infection.


Asunto(s)
Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Adulto , Aminohidrolasas/genética , Aminohidrolasas/inmunología , Antivirales/inmunología , Factores de Restricción Antivirales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Estudios de Casos y Controles , Estudios de Cohortes , Estudios Transversales , Progresión de la Enfermedad , Elafina/genética , Elafina/inmunología , Femenino , Genitales Femeninos/inmunología , Infecciones por VIH/virología , Sobrevivientes de VIH a Largo Plazo , Seronegatividad para VIH/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Tejido Linfoide/inmunología , Masculino , Persona de Mediana Edad , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Mucosa Bucal/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleasas/genética , Ribonucleasas/inmunología , Proteína 1 que Contiene Dominios SAM y HD , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Replicación Viral/inmunología , Adulto Joven , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/inmunología
10.
J Immunol ; 195(1): 339-46, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26026060

RESUMEN

The mammalian target of rapamycin (mTOR) signaling pathway integrates environmental cues to regulate cell growth and survival through various mechanisms. However, how mTORC1 responds to acute inflammatory signals to regulate bowel regeneration is still obscure. In this study, we investigated the role of mTORC1 in acute inflammatory bowel disease. Inhibition of mTORC1 activity by rapamycin treatment or haploinsufficiency of Rheb through genetic modification in mice impaired intestinal cell proliferation and induced cell apoptosis, leading to high mortality in dextran sodium sulfate- and 2,4,6-trinitrobenzene sulfonic acid-induced colitis models. Through bone marrow transplantation, we found that mTORC1 in nonhematopoietic cells played a major role in protecting mice from colitis. Reactivation of mTORC1 activity by amino acids had a positive therapeutic effect in mTORC1-deficient Rheb(+/-) mice. Mechanistically, mTORC1 mediated IL-6-induced Stat3 activation in intestinal epithelial cells to stimulate the expression of downstream targets essential for cell proliferation and tissue regeneration. Therefore, mTORC1 signaling critically protects against inflammatory bowel disease through modulation of inflammation-induced Stat3 activity. As mTORC1 is an important therapeutic target for multiple diseases, our findings will have important implications for the clinical usage of mTORC1 inhibitors in patients with acute inflammatory bowel disease.


Asunto(s)
Colitis/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Complejos Multiproteicos/antagonistas & inhibidores , Neuropéptidos/inmunología , Factor de Transcripción STAT3/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Trasplante de Médula Ósea , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/genética , Colitis/mortalidad , Regulación de la Expresión Génica , Haploinsuficiencia , Humanos , Interleucina-6/genética , Interleucina-6/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/deficiencia , Proteínas de Unión al GTP Monoméricas/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Neuropéptidos/deficiencia , Neuropéptidos/genética , Proteína Homóloga de Ras Enriquecida en el Cerebro , Factor de Transcripción STAT3/genética , Transducción de Señal , Dodecil Sulfato de Sodio , Análisis de Supervivencia , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Ácido Trinitrobencenosulfónico
11.
J Immunol ; 194(8): 3852-60, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25786685

RESUMEN

Leishmania major is a parasite that resides and replicates in macrophages. We previously showed that the parasite enhanced CD40-induced Raf-MEK-ERK signaling but inhibited PI3K-MKK-p38MAPK signaling to proleishmanial effects. As Raf and PI3K have a Ras-binding domain but exert opposite effects on Leishmania infection, we examined whether Ras isoforms had differential roles in Leishmania infection. We observed that L. major enhanced N-Ras and H-Ras expression but inhibited K-Ras expression in macrophages. L. major infection enhanced N-Ras activity but inhibited H-Ras and K-Ras activity. TLR2 short hairpin RNA or anti-TLR2 or anti-lipophosphoglycan Abs reversed the L. major-altered N-Ras and K-Ras expressions. Pam3CSK4, a TLR2 ligand, enhanced N-Ras expression but reduced K-Ras expression, indicating TLR2-regulated Ras expression in L. major infection. Whereas N-Ras silencing reduced L. major infection, K-Ras and H-Ras silencing enhanced the infection both in macrophages in vitro and in C57BL/6 mice. BALB/c-derived macrophages transduced with lentivirally expressed N-Ras short hairpin RNA and pulsed with L. major-expressed MAPK10 enhanced MAPK10-specific Th1-type response. CD40-deficient mice primed with these macrophages had reduced L. major infection, accompanied by higher IFN-γ but less IL-4 production. As N-Ras is activated by Sos, a guanine nucleotide exchange factor, we modeled the N-Ras-Sos interaction and designed two peptides from their interface. Both the cell-permeable peptides reduced L. major infection in BALB/c mice but not in CD40-deficient mice. These data reveal the L. major-enhanced CD40-induced N-Ras activation as a novel immune evasion strategy and the potential for Ras isoform-targeted antileishmanial immunotherapy and immunoprophylaxis.


Asunto(s)
Antígenos CD40/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Animales , Antígenos CD40/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Evasión Inmune/efectos de los fármacos , Evasión Inmune/genética , Evasión Inmune/inmunología , Inmunoterapia , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/patología , Leishmaniasis Cutánea/prevención & control , Lipopéptidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Proteína Quinasa 10 Activada por Mitógenos/genética , Proteína Quinasa 10 Activada por Mitógenos/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas de Unión al GTP Monoméricas/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Proteína Son Of Sevenless Drosofila/genética , Proteína Son Of Sevenless Drosofila/inmunología , Células TH1/inmunología , Células TH1/patología , Receptor Toll-Like 2 , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
12.
J Immunol ; 194(8): 3623-33, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25769924

RESUMEN

Aicardi-Goutières syndrome (AGS) is a monogenic inflammatory encephalopathy caused by mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, or MDA5. Mutations in those genes affect normal RNA/DNA intracellular metabolism and detection, triggering an autoimmune response with an increase in cerebral IFN-α production by astrocytes. Microangiopathy and vascular disease also contribute to the neuropathology in AGS. In this study, we report that AGS gene silencing of TREX1, SAMHD1, RNASEH2A, and ADAR1 by short hairpin RNAs in human neural stem cell-derived astrocytes, human primary astrocytes, and brain-derived endothelial cells leads to an antiviral status of these cells compared with nontarget short hairpin RNA-treated cells. We observed a distinct activation of the IFN-stimulated gene signature with a substantial increase in the release of proinflammatory cytokines (IL-6) and chemokines (CXCL10 and CCL5). A differential impact of AGS gene silencing was noted; silencing TREX1 gave rise to the most dramatic in both cell types. Our findings fit well with the observation that patients carrying mutations in TREX1 experience an earlier onset and fatal outcome. We provide in the present study, to our knowledge for the first time, insight into how astrocytic and endothelial activation of antiviral status may differentially lead to cerebral pathology, suggesting a rational link between proinflammatory mediators and disease severity in AGS.


Asunto(s)
Astrocitos/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Citocinas/inmunología , Células Endoteliales/inmunología , Interferón-alfa/inmunología , Malformaciones del Sistema Nervioso/inmunología , Células-Madre Neurales/inmunología , Adenosina Desaminasa/genética , Adenosina Desaminasa/inmunología , Astrocitos/patología , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/mortalidad , Enfermedades Autoinmunes del Sistema Nervioso/patología , Citocinas/genética , Células Endoteliales/patología , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/inmunología , Silenciador del Gen , Células HEK293 , Humanos , Interferón-alfa/genética , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Mutación , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/mortalidad , Malformaciones del Sistema Nervioso/patología , Células-Madre Neurales/patología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Ribonucleasa H/genética , Ribonucleasa H/inmunología , Proteína 1 que Contiene Dominios SAM y HD
13.
J Virol ; 89(2): 1329-39, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392215

RESUMEN

UNLABELLED: In order to develop strategies to prevent HIV-1 (human immunodeficiency virus type 1) transmission, it is crucial to better characterize HIV-1 target cells in the female reproductive tract (FRT) mucosae and to identify effective innate responses. Control of HIV-1 infection in the decidua (the uterine mucosa during pregnancy) can serve as a model to study natural mucosal protection. Macrophages are the main HIV-1 target cells in the decidua. Here we report that in vitro, macrophages and T cells are the main HIV-1 targets in the endometrium in nonpregnant women. As reported for decidual macrophages (dM), endometrial macrophages (eM) were found to have an M2-like phenotype (CD68+ CD163+ CD206+ IL-10high). However, eM and dM may belong to different subpopulations, as they differently express certain markers and secrete different amounts of proinflammatory and anti-inflammatory cytokines. We observed strong expression of the SAMHD1 restriction factor and weak expression of its inactive form (pSAMHD1, phosphorylated at residue Thr592) in both eM and dM. Infection of macrophages from both tissues was enhanced in the presence of the viral protein Vpx, suggesting a role for SAMHD1 in the restriction of HIV-1 infection. This study and further comparisons of the decidua with FRT mucosae in nonpregnant women should help to identify mechanisms of mucosal protection against HIV-1 infection. IMPORTANCE: The female reproductive tract mucosae are major portals of HIV-1 entry into the body. The decidua (uterine mucosa during pregnancy) can serve as a model for studying natural mucosal protection against HIV-1 transmission. A comparison of target cells and innate responses in the decidua versus the endometrium in nonpregnant women could help to identify protective mechanisms. Here, we report for the first time that macrophages are one of the main HIV-1 target cells in the endometrium and that infection of macrophages from both the endometrium and the decidua is restricted by SAMHD1. These findings might have implications for the development of vaccines to prevent HIV-1 mucosal transmission.


Asunto(s)
Endometrio/inmunología , VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Macrófagos/inmunología , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/metabolismo , Adulto , Antígenos CD/análisis , Femenino , Humanos , Inmunofenotipificación , Interleucina-10/análisis , Macrófagos/química , Persona de Mediana Edad , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD , Linfocitos T/inmunología , Linfocitos T/virología , Adulto Joven
14.
Monoclon Antib Immunodiagn Immunother ; 33(5): 344-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25358004

RESUMEN

The sterile alpha motif and HD domain 1 (SAMHD1) protein has been identified as a novel innate immunity restriction factor that participates in processes crucial to the viral life cycle. In the present study, we describe a procedure to generate monoclonal antibodies (MAbs) against porcine SAMHD1 and investigate its characteristics to analyze the expression of endogenous SAMHD1. The open reading frame of porcine SAMHD1 was cloned into the prokaryotic expression vector pCold-TF DNA to construct a recombinant plasmid pcold-pSAMHD1 and induce expression of recombinant porcine SAMHD1 protein by IPTG in Escherichia coli Rosetta. The purified recombinant porcine SAMHD1 protein was used to prepare MAbs of SAMHD1. After subcloning five times hybridoma cell clones expressing SAMHD1, MAbs were generated. Western blot analysis and indirect immunofluorescence assay showed that the overexpressed porcine SAMHD1 in 293T cells and endogenous SAMHD1 protein in porcine cell lines could be specifically recognized by the MAbs produced in this study. In conclusion, specific MAbs of porcine SAMHD1 are reported, and these MAbs provide a valuable tool for further studies of SAMHD1-mediated signaling in virus-infected cells to elucidate the underlying antiviral mechanism.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Escherichia coli/genética , Células HEK293 , Células HeLa , Humanos , Hibridomas/inmunología , Proteínas de Unión al GTP Monoméricas/genética , Sistemas de Lectura Abierta/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Porcinos
15.
Cytokine Growth Factor Rev ; 25(6): 657-68, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25240798

RESUMEN

Retroviruses can selectively trigger an array of innate immune responses through various PRR. The identification and the characterization of the molecular basis of retroviral DNA sensing by the DNA sensors IFI16 and cGAS has been one of the most exciting developments in viral immunology in recent years. DNA sensing by these cytosolic sensors not only leads to the initiation of the type I interferon (IFN) antiviral response and the induction of the inflammatory response, but also triggers cell death mechanisms including pyroptosis and apoptosis in retrovirus-infected cells, thereby providing important insights into the pathophysiology of chronic retroviral infection. Host restriction factors such as SAMHD1 and Trex1 play important roles in regulating innate immune sensing, and have led to the idea that innate immune defense and host restriction actually converge at different levels to determine the outcome of retroviral infection. In this review, we discuss the sensing of retroviruses by cytosolic DNA sensors, the relevance of host factors during retroviral infection, and the interplay between host factors and the innate antiviral response in different cell types, within the context of two human pathogenic retroviruses - human immunodeficiency virus (HIV-1) and human T cell-leukemia virus type I (HTLV-1).


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Infecciones por HTLV-I/inmunología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Inmunidad Innata , Animales , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/inmunología , Infecciones por VIH/genética , VIH-1/genética , Infecciones por HTLV-I/genética , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
16.
Dev Comp Immunol ; 47(2): 234-46, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25106914

RESUMEN

The sterile alpha motif and HD domain 1 (SAMHD1) protein is a novel innate immunity restriction factor that inhibits HIV-1 infection in myeloid cells. Here, we cloned the full-length SAMHD1 complementary DNA (cDNA) from porcine peripheral blood lymphocytes. The porcine SAMHD1 cDNA was of 3951 bp with an open reading frame of 1884 bp, encoding a polypeptide of 627 amino acids. Porcine SAMHD1 mRNA was detected in all swine tissues examined, with the higher expression in the tonsil, lung, liver, and lymph node tissues. The SAMHD1 protein was localized to the nucleus. Overexpression of SAMHD1 blocked the proliferation of HuN4, a highly pathogenic strain of porcine reproductive and respiratory syndrome virus (HP-PRRSV), in MARC-145 cells, by inhibiting the synthesis of the HuN4 complement RNA. The antiviral effects of the simian SAMHD1 protein were nearly equivalent to those of porcine SAMHD1 in the HuN4-infected MARC-145 cells. Phosphorylation analysis of SAMHD1 showed that overexpressed SAMHD1 protein was in primarily an unphosphorylated state. SAMHD1 overexpression increased the transcript abundance of IFN-stimulated genes ISG15 and ISG56. The mRNA levels of SAMHD1 and ISGs were significantly increased in porcine alveolar macrophages infected with HP-PRRSV. SAMHD1 protein level was also elevated, and the protein was not phosphorylated during infection. Collectively, our data indicate that SAMHDI inhibits HP-PRRSV proliferation through inhibiting the replication of HP-PRRSV. SAMHD1 might be the protein participating in the IFN signaling and is thus an important immunoregulatory protein in innate immunity.


Asunto(s)
Clonación Molecular , Macrófagos Alveolares/inmunología , Proteínas de Unión al GTP Monoméricas/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular , Núcleo Celular/inmunología , Núcleo Celular/virología , Citocinas/genética , Citocinas/inmunología , ADN Complementario/genética , ADN Complementario/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Macrófagos Alveolares/patología , Macrófagos Alveolares/virología , Datos de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/genética , Sistemas de Lectura Abierta , Fosforilación , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/patología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Porcinos , Carga Viral , Replicación Viral
17.
Artículo en Ruso | MEDLINE | ID: mdl-25051707

RESUMEN

Main scientific-research studies regarding protein bacterial toxins of the most widespread bacteria that belong to Staphylococcus spp. genus and in particular the most pathogenic species for humans--Staphylococcus aureus, are analyzed. Structural and biological properties of protein toxins that have received the name of staphylococcus pyrogenic toxins (PTSAg) are presented. Data regarding genetic regulation of secretion and synthesis of these toxins and 3 main regulatory genetic systems (agr--accessory gene regulator, xpr--extracellular protein regulator, sar--staphylococcal accessory regulator) that coordinate synthesis of the most important protein toxins and enzymes for virulence of S. aureus, are presented.


Asunto(s)
Toxinas Bacterianas/inmunología , Regulación Bacteriana de la Expresión Génica/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Linfocitos T/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/clasificación , Toxinas Bacterianas/genética , Anergia Clonal , Transferencia de Gen Horizontal , Interacciones Huésped-Patógeno , Humanos , Activación de Linfocitos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Superantígenos/genética , Superantígenos/inmunología , Linfocitos T/microbiología , Linfocitos T/patología , Transactivadores/genética , Transactivadores/inmunología , Virulencia
18.
Proc Natl Acad Sci U S A ; 111(18): E1843-51, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753578

RESUMEN

The HIV-1 restriction factor sterile α-motif/histidine-aspartate domain-containing protein 1 (SAMHD1) is a tetrameric protein that catalyzes the hydrolysis of all dNTPs to the deoxynucleoside and tripolyphosphate, which effectively depletes the dNTP substrates of HIV reverse transcriptase. Here, we establish that SAMHD1 is activated by GTP binding to guanine-specific activator sites (A1) as well as coactivation by substrate dNTP binding to a distinct set of nonspecific activator sites (A2). Combined activation by GTP and dNTPs results in a long-lived tetrameric form of SAMHD1 that persists for hours, even after activating nucleotides are withdrawn from the solution. These results reveal an ordered model for assembly of SAMHD1 tetramer from its inactive monomer and dimer forms, where GTP binding to the A1 sites generates dimer and dNTP binding to the A2 and catalytic sites generates active tetramer. Thus, cellular regulation of active SAMHD1 is not determined by GTP alone but instead, the levels of all dNTPs and the generation of a persistent tetramer that is not in equilibrium with free activators. The significance of the long-lived activated state is that SAMHD1 can remain active long after dNTP pools have been reduced to a level that would lead to inactivation. This property would be important in resting CD4(+) T cells, where dNTP pools are reduced to nanomolar levels to restrict infection by HIV-1.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Guanosina Trifosfato/metabolismo , VIH-1/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Dominio Catalítico , Nucleótidos de Desoxiguanina/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Nucleótidos de Desoxiuracil/metabolismo , Activación Enzimática , VIH-1/inmunología , VIH-1/patogenicidad , Humanos , Inmunidad Innata , Cinética , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/inmunología , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Proteína 1 que Contiene Dominios SAM y HD , Especificidad por Sustrato , Tionucleósidos/metabolismo
19.
J Virol ; 88(9): 4976-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24554663

RESUMEN

UNLABELLED: CD4(+) and CD8(+) memory T cells with stem cell-like properties (T(SCM) cells) have been identified in mice, humans, and nonhuman primates and are being investigated for antitumor and antiviral vaccines and immunotherapies. Whether CD4(+) T(SCM) cells are infected by human immunodeficiency virus (HIV) was investigated by using a combination HIV reporter virus system in vitro and by direct staining for HIV p24 antigen ex vivo. A proportion of T(SCM) cells were found to express the HIV coreceptors CCR5 and CXCR4 and were infected by HIV both in vitro and in vivo. Analysis of viral outcome following fusion using the combination reporter virus system revealed that T(SCM) cells can become productively or latently infected, although the vast majority of T(SCM) cells are abortively infected. Knockdown of the HIV restriction factor SAMHD1 using Vpx-containing simian immunodeficiency virus (SIV) virion-like particles enhanced the productive infection of T(SCM) cells, indicating that SAMHD1 contributes to abortive infection in these cells. These results demonstrate that CD4(+) T(SCM) cells are targets for HIV infection, that they become productively or latently infected at low levels, and that SAMHD1 expression promotes abortive infection of this important memory cell subset. IMPORTANCE: Here we demonstrate the susceptibility of CD4(+) memory stem cells (T(SCM) cells) to infection by HIV in vitro and in vivo, provide an in-depth analysis of coreceptor expression, demonstrate the infection of naïve and memory CD4(+) T cell subsets with both CCR5- and CXCR4-tropic HIV, and also perform outcome analysis to calculate the percentage of cells that are productively, latently, or abortively infected. Through these outcome studies, we determined that the vast majority of T(SCM) cells are abortively infected by HIV, and we demonstrate that knockdown of SAMHD1 significantly increases the frequency of infection of this CD4(+) T cell subset, indicating that SAMHD1 is an active restriction factor in T(SCM) cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , VIH-1/crecimiento & desarrollo , Proteínas de Unión al GTP Monoméricas/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Células Madre/virología , Expresión Génica , Voluntarios Sanos , Humanos , Receptores CCR5/biosíntesis , Receptores CXCR4/biosíntesis , Receptores del VIH/biosíntesis , Proteína 1 que Contiene Dominios SAM y HD
20.
J Infect Dis ; 210(1): 89-98, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24403554

RESUMEN

miR-155 (microRNA-155) is an important noncoding RNA in regulating host inflammatory responses. However, its regulatory role in ocular infection remains unclear. Our study first explored the function of miR-155 in Pseudomonas aeruginosa-induced keratitis, one of the most common sight-threatening ocular diseases. We found that miR-155 expression was enhanced in human and mouse corneas after P. aeruginosa infection and was mainly expressed in macrophages but not neutrophils. In vivo studies demonstrated that miR-155 knockout mice displayed more resistance to P. aeruginosa keratitis, with a higher inducible nitric oxide synthase level and a lower bacterial burden. More importantly, in vitro data indicated that miR-155 suppressed the macrophage-mediated bacterial phagocytosis and intracellular killing of P. aeruginosa by targeting Rheb (Ras homolog enriched in brain). To the best of our knowledge, this is the first study to explore the role of miR-155 in bacterial keratitis, which may provide a promising target for clinical treatment of P. aeruginosa keratitis and other infectious diseases.


Asunto(s)
Queratitis/inmunología , Queratitis/microbiología , MicroARNs/metabolismo , Proteínas de Unión al GTP Monoméricas/inmunología , Neuropéptidos/inmunología , Pseudomonas aeruginosa/inmunología , Adulto , Animales , Carga Bacteriana , Femenino , Humanos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Viabilidad Microbiana , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Fagocitosis , Proteína Homóloga de Ras Enriquecida en el Cerebro , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA