Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nature ; 583(7818): 845-851, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32699415

RESUMEN

Malignant transformation of cells typically involves several genetic lesions, whose combined activity gives rise to cancer1. Here we analyse 1,148 patient-derived B-cell leukaemia (B-ALL) samples, and find that individual mutations do not promote leukaemogenesis unless they converge on one single oncogenic pathway that is characteristic of the differentiation stage of transformed B cells. Mutations that are not aligned with this central oncogenic driver activate divergent pathways and subvert transformation. Oncogenic lesions in B-ALL frequently mimic signalling through cytokine receptors at the pro-B-cell stage (via activation of the signal-transduction protein STAT5)2-4 or pre-B-cell receptors in more mature cells (via activation of the protein kinase ERK)5-8. STAT5- and ERK-activating lesions are found frequently, but occur together in only around 3% of cases (P = 2.2 × 10-16). Single-cell mutation and phospho-protein analyses reveal the segregation of oncogenic STAT5 and ERK activation to competing clones. STAT5 and ERK engage opposing biochemical and transcriptional programs that are orchestrated by the transcription factors MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway comes at the expense of the principal oncogenic driver and reverses transformation. Conversely, deletion of divergent pathway components accelerates leukaemogenesis. Thus, persistence of divergent signalling pathways represents a powerful barrier to transformation, while convergence on one principal driver defines a central event in leukaemia initiation. Pharmacological reactivation of suppressed divergent circuits synergizes strongly with inhibition of the principal oncogenic driver. Hence, reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to enhance treatment responses.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Transformación Celular Neoplásica , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Transducción de Señal , Animales , Linfocitos B/patología , Línea Celular Tumoral , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Transcripción STAT5/metabolismo
2.
Nature ; 588(7838): 491-497, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33149299

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) has previously been identified as an endosomal protein that blocks viral infection1-3. Here we studied clinical cohorts of patients with B cell leukaemia and lymphoma, and identified IFITM3 as a strong predictor of poor outcome. In normal resting B cells, IFITM3 was minimally expressed and mainly localized in endosomes. However, engagement of the B cell receptor (BCR) induced both expression of IFITM3 and phosphorylation of this protein at Tyr20, which resulted in the accumulation of IFITM3 at the cell surface. In B cell leukaemia, oncogenic kinases phosphorylate IFITM3 at Tyr20, which causes constitutive localization of this protein at the plasma membrane. In a mouse model, Ifitm3-/- naive B cells developed in normal numbers; however, the formation of germinal centres and the production of antigen-specific antibodies were compromised. Oncogenes that induce the development of leukaemia and lymphoma did not transform Ifitm3-/- B cells. Conversely, the phosphomimetic IFITM3(Y20E) mutant induced oncogenic PI3K signalling and initiated the transformation of premalignant B cells. Mechanistic experiments revealed that IFITM3 functions as a PIP3 scaffold and central amplifier of PI3K signalling. The amplification of PI3K signals depends on IFITM3 using two lysine residues (Lys83 and Lys104) in its conserved intracellular loop as a scaffold for the accumulation of PIP3. In Ifitm3-/- B cells, lipid rafts were depleted of PIP3, which resulted in the defective expression of over 60 lipid-raft-associated surface receptors, and impaired BCR signalling and cellular adhesion. We conclude that the phosphorylation of IFITM3 that occurs after B cells encounter antigen induces a dynamic switch from antiviral effector functions in endosomes to a PI3K amplification loop at the cell surface. IFITM3-dependent amplification of PI3K signalling, which in part acts downstream of the BCR, is critical for the rapid expansion of B cells with high affinity to antigen. In addition, multiple oncogenes depend on IFITM3 to assemble PIP3-dependent signalling complexes and amplify PI3K signalling for malignant transformation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Animales , Antígenos CD19/metabolismo , Linfocitos B/enzimología , Linfocitos B/inmunología , Linfocitos B/patología , Transformación Celular Neoplásica , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/patología , Humanos , Integrinas/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Modelos Moleculares , Fosforilación , Receptores de Antígenos de Linfocitos B/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33531346

RESUMEN

Unlike other cell types, developing B cells undergo multiple rounds of somatic recombination and hypermutation to evolve high-affinity antibodies. Reflecting the high frequency of DNA double-strand breaks, adaptive immune protection by B cells comes with an increased risk of malignant transformation. B lymphoid transcription factors (e.g., IKZF1 and PAX5) serve as metabolic gatekeepers by limiting glucose to levels insufficient to fuel transformation. We here identified aberrant expression of the lactonase PON2 in B cell acute lymphoblastic leukemia (B-ALL) as a mechanism to bypass metabolic gatekeeper functions. Compared to normal pre-B cells, PON2 expression was elevated in patient-derived B-ALL samples and correlated with poor clinical outcomes in pediatric and adult cohorts. Genetic deletion of Pon2 had no measurable impact on normal B cell development. However, in mouse models for BCR-ABL1 and NRASG12D-driven B-ALL, deletion of Pon2 compromised proliferation, colony formation, and leukemia initiation in transplant recipient mice. Compromised leukemogenesis resulted from defective glucose uptake and adenosine triphosphate (ATP) production in PON2-deficient murine and human B-ALL cells. Mechanistically, PON2 enabled glucose uptake by releasing the glucose-transporter GLUT1 from its inhibitor stomatin (STOM) and genetic deletion of STOM largely rescued PON2 deficiency. While not required for glucose transport, the PON2 lactonase moiety hydrolyzes the lactone-prodrug 3OC12 to form a cytotoxic intermediate. Mirroring PON2 expression levels in B-ALL, 3OC12 selectively killed patient-derived B-ALL cells but was well tolerated in transplant recipient mice. Hence, while B-ALL cells critically depend on aberrant PON2 expression to evade metabolic gatekeeper functions, PON2 lactonase activity can be leveraged as synthetic lethality to overcome drug resistance in refractory B-ALL.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Linfocitos B/metabolismo , Carcinogénesis/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Arildialquilfosfatasa/genética , Carcinogénesis/genética , Línea Celular Tumoral , Células Cultivadas , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Unión Proteica
5.
Cell Physiol Biochem ; 54(4): 767-790, 2020 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-32830930

RESUMEN

The pandemic of the severe acute respiratory syndrome coronavirus (SARS-CoV)-2 at the end of 2019 marked the third outbreak of a highly pathogenic coronavirus affecting the human population in the past twenty years. Cross-species zoonotic transmission of SARS-CoV-2 has caused severe pathogenicity and led to more than 655,000 fatalities worldwide until July 28, 2020. Outbursts of this virus underlined the importance of controlling infectious pathogens across international frontiers. Unfortunately, there is currently no clinically approved antiviral drug or vaccine against SARS-CoV-2, although several broad-spectrum antiviral drugs targeting multiple RNA viruses have shown a positive response and improved recovery in patients. In this review, we compile our current knowledge of the emergence, transmission, and pathogenesis of SARS-CoV-2 and explore several features of SARS-CoV-2. We emphasize the current therapeutic approaches used to treat infected patients. We also highlight the results of in vitro and in vivo data from several studies, which have broadened our knowledge of potential drug candidates for the successful treatment of patients infected with and discuss possible virus and host-based treatment options against SARS-CoV-2.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus , Pandemias , Neumonía Viral , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/genética , Betacoronavirus/fisiología , COVID-19 , Vacunas contra la COVID-19 , Coronaviridae/patogenicidad , Infecciones por Coronaviridae/epidemiología , Infecciones por Coronaviridae/virología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/terapia , Infecciones por Coronavirus/transmisión , Síndrome de Liberación de Citoquinas/etiología , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos , Endocitosis/efectos de los fármacos , Predicción , Genoma Viral , Salud Global , Humanos , Inmunidad Colectiva , Inmunización Pasiva , Pandemias/prevención & control , Péptido Hidrolasas/farmacología , Péptido Hidrolasas/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/epidemiología , Neumonía Viral/prevención & control , Neumonía Viral/transmisión , ARN Viral/genética , Receptores de Coronavirus , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas Virales , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Zoonosis , Tratamiento Farmacológico de COVID-19 , Sueroterapia para COVID-19
6.
Cell Physiol Biochem ; 42(4): 1358-1365, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28704812

RESUMEN

BACKGROUND: Cellular glucose uptake may involve either non-concentrative glucose carriers of the GLUT family or Na+-coupled glucose-carrier SGLT1, which accumulates glucose against glucose gradients and may thus accomplish cellular glucose uptake even at dramatically decreased extracellular glucose concentrations. SGLT1 is not only expressed in epithelia but as well in tumour cells and immune cells. Immune cell functions strongly depend on their metabolism, therefore we hypothesized that deficiency of SGLT1 modulates the defence against bacterial infection. To test this hypothesis, we infected wild type mice and gene targeted mice lacking functional SGLT1 with Listeria monocytogenes. METHODS: SGLT1 deficient mice and wild type littermates were infected with 1x104 CFU Listeria monocytogenes intravenously. Bacterial titers were determined by colony forming assay, SGLT1, TNF-α, IL-6 and IL-12a transcript levels were determined by qRT-PCR, as well as SGLT1 protein abundance and localization by immunohistochemistry. RESULTS: Genetic knockout of SGLT1 (Slc5a1-/- mice) significantly compromised bacterial clearance following Listeria monocytogenes infection with significantly enhanced bacterial load in liver, spleen, kidney and lung, and significantly augmented hepatic expression of TNF-α and IL-12a. While all wild type mice survived, all SGLT1 deficient mice died from the infection. CONCLUSIONS: SGLT1 is required for bacterial clearance and host survival following murine Listeria infection.


Asunto(s)
Expresión Génica/inmunología , Glucosa/inmunología , Listeriosis/genética , Listeriosis/mortalidad , Hígado/inmunología , Transportador 1 de Sodio-Glucosa/inmunología , Animales , Carga Bacteriana , Transporte Biológico , Glucosa/metabolismo , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Riñón/inmunología , Riñón/microbiología , Riñón/patología , Listeria monocytogenes , Listeriosis/inmunología , Listeriosis/microbiología , Hígado/microbiología , Hígado/patología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/inmunología , Transportador 1 de Sodio-Glucosa/deficiencia , Transportador 1 de Sodio-Glucosa/genética , Bazo/inmunología , Bazo/microbiología , Bazo/patología , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
7.
Cell Physiol Biochem ; 39(3): 1209-28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27595398

RESUMEN

BACKGROUND: Similar to tumor cells, activated T-lymphocytes generate ATP mainly by glycolytic degradation of glucose. Lymphocyte glucose uptake involves non-concentrative glucose carriers of the GLUT family. In contrast to GLUT isoforms, Na+-coupled glucose-carrier SGLT1 accumulates glucose against glucose gradients and is effective at low extracellular glucose concentrations. The present study explored expression and regulation of SGLT1 in activated murine splenic cytotoxic T cells (CTLs) and human Jurkat T cells. METHODS: FACS analysis, immunofluorescence, confocal microscopy, chemiluminescence and Western blotting were employed to estimate SGLT1 expression, function and regulation in lymphocytes, as well as dual electrode voltage clamp in SGLT1 ± JAK3 expressing Xenopus oocytes to quantify the effect of janus kinase3 (JAK3) on SGLT1 function. RESULTS: SGLT1 is expressed in murine CTLs and also in human Jurkat T cells. 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose uptake was significantly decreased by SGLT1-blocker phloridzin (0.2 mM) and by pharmacological inhibition of JAK3 with WHI-P131 (156 µM), WHI-P154 (11.2 µM) and JAK3 inhibitor VI (0.5 µM). Electrogenic glucose transport (Iglucose) in Xenopus oocytes expressing human SGLT1 was increased by additional expression of human wild type JAK3, active A568VJAK3 but not inactive K851AJAK3. Coexpression of JAK3 enhanced the maximal transport rate without significantly modifying affinity of the carrier. Iglucose in SGLT1+JAK3 expressing oocytes was significantly decreased by WHI-P154 (11.2 µM). JAK3 increased the SGLT1 protein abundance in the cell membrane. Inhibition of carrier insertion by brefeldin A (5 µM) in SGLT1+JAK3 expressing oocytes resulted in a decline of Iglucose, which was similar in presence and absence of JAK3. CONCLUSIONS: SGLT1 is expressed in murine cytotoxic T cells and human Jurkat T cells and significantly contributes to glucose uptake in those cells post activation. JAK3 up-regulates SGLT1 activity by increasing the carrier protein abundance in the cell membrane, an effect enforcing cellular glucose uptake into activated lymphocytes and thus contributing to the immune response.


Asunto(s)
Glucosa/inmunología , Janus Quinasa 3/genética , Oocitos/metabolismo , Transportador 1 de Sodio-Glucosa/genética , Linfocitos T Citotóxicos/inmunología , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/farmacología , Animales , Transporte Biológico , Brefeldino A/farmacología , Células CACO-2 , Desoxiglucosa/análogos & derivados , Desoxiglucosa/farmacología , Regulación de la Expresión Génica , Glucosa/farmacología , Humanos , Janus Quinasa 3/inmunología , Células Jurkat , Activación de Linfocitos , Ratones , Oocitos/citología , Oocitos/efectos de los fármacos , Técnicas de Placa-Clamp , Florizina/farmacología , Cultivo Primario de Células , Quinazolinas/farmacología , Transducción de Señal , Transportador 1 de Sodio-Glucosa/inmunología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Transgenes , Xenopus laevis
8.
Cell Physiol Biochem ; 38(4): 1343-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27007114

RESUMEN

BACKGROUND: Graft versus host disease (GvHD) occurs in 20% of cases with patients having an MHC I matched bone marrow transplantation (BMT). Mechanisms causing this disease remain to be studied. METHODS: Here we used a CD8+ T cell transgenic mouse line (P14/CD45.1+) and transgenic DEE mice bearing ubiquitously the glycoprotein 33-41 (GP33) antigen derived from the major lymphocytic choriomeningitis virus (LCMV) epitope to study mechanisms of tolerance in anti-host reactive CD8+ T cells after BMT. RESULTS: We found that anti-host reactive CD8+ T cells (P14 T cells) were not negatively selected in the thymus and that they were present in wild type (WT) recipient mice as well as in DEE recipient mice. Anti-host reactive CD8+ T cells ignored the GP33 antigen expressed ubiquitously by host cells but they could be activated ex vivo via LCMV-infection. Lipopolysaccharides (LPS) induced transient cell damage in DEE mice bearing anti-host reactive CD8+ T cells after BMT, suggesting that induction of host inflammatory response could break antigen ignorance. Introducing the GP33 antigen into BM cells led to deletion of anti-host reactive CD8+ T cells. CONCLUSION: We found that after BMT anti-host reactive CD8+ T cells ignored host antigen in recipients and that they were only deleted when host antigen was present in hematopoietic cells. Moreover, LPS-induced immune activation contributed to induction of alloreactivity of anti-host reactive CD8+ T cells after BMT.


Asunto(s)
Trasplante de Médula Ósea , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Alanina Transaminasa/metabolismo , Animales , Anticuerpos/inmunología , Antígenos Virales/genética , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Epítopos/inmunología , Citometría de Flujo , Glicoproteínas/genética , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lipopolisacáridos/toxicidad , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Trasplante Homólogo , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
9.
J Virol ; 89(9): 4748-59, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25673724

RESUMEN

UNLABELLED: The B cell-activating factor (BAFF) is critical for B cell development and humoral immunity in mice and humans. While the role of BAFF in B cells has been widely described, its role in innate immunity remains unknown. Using BAFF receptor (BAFFR)-deficient mice, we characterized BAFFR-related innate and adaptive immune functions following infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). We identified a critical role for BAFFR signaling in the generation and maintenance of the CD169(+) macrophage compartment. Consequently, Baffr(-) (/) (-) mice exhibited limited induction of innate type I interferon production after viral infection. Lack of BAFFR signaling reduced virus amplification and presentation following viral infection, resulting in highly reduced antiviral adaptive immune responses. As a consequence, BAFFR-deficient mice showed exacerbated and fatal disease after viral infection. Mechanistically, transient lack of B cells in Baffr(-) (/) (-) animals resulted in limited lymphotoxin expression, which is critical for maintenance of CD169(+) cells. In conclusion, BAFFR signaling affects both innate and adaptive immune activation during viral infections. IMPORTANCE: Viruses cause acute and chronic infections in humans resulting in millions of deaths every year. Innate immunity is critical for the outcome of a viral infection. Innate type I interferon production can limit viral replication, while adaptive immune priming by innate immune cells induces pathogen-specific immunity with long-term protection. Here, we show that BAFFR deficiency not only perturbed B cells, but also resulted in limited CD169(+) macrophages. These macrophages are critical in amplifying viral particles to trigger type I interferon production and initiate adaptive immune priming. Consequently, BAFFR deficiency resulted in reduced enforced viral replication, limited type I interferon production, and reduced adaptive immunity compared to BAFFR-competent controls. As a result, BAFFR-deficient mice were predisposed to fatal viral infections. Thus, BAFFR expression is critical for innate immune activation and antiviral immunity.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Macrófagos/química , Macrófagos/inmunología , Receptores de Interleucina-4/deficiencia , Infecciones por Rhabdoviridae/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/análisis , Inmunidad Adaptativa , Animales , Inmunidad Innata , Interferón Tipo I/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones Noqueados , Transducción de Señal , Vesiculovirus/inmunología
10.
J Autoimmun ; 67: 82-89, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26553386

RESUMEN

The induction of innate and adaptive immunity is essential for controlling viral infections. Limited or overwhelming innate immunity can negatively impair the adaptive immune response. Therefore, balancing innate immunity separately from activating the adaptive immune response would result in a better antiviral immune response. Recently, we demonstrated that Usp18-dependent replication of virus in secondary lymphatic organs contributes to activation of the innate and adaptive immune responses. Whether specific mechanisms can balance innate and adaptive immunity separately remains unknown. In this study, using lymphocytic choriomeningitis virus (LCMV) and replication-deficient single-cycle LCMV vectors, we found that viral replication of the initial inoculum is essential for activating virus-specific CD8(+) T cells. In contrast, extracellular distribution of virus along the splenic conduits is necessary for inducing systemic levels of type I interferon (IFN-I). Although enforced virus replication is driven primarily by Usp18, B cell-derived lymphotoxin beta contributes to the extracellular distribution of virus along the splenic conduits. Therefore, lymphotoxin beta regulates IFN-I induction independently of CD8(+) T-cell activity. We found that two separate mechanisms act together in the spleen to guarantee amplification of virus during infection, thereby balancing the activation of the innate and adaptive immune system.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Replicación Viral/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/biosíntesis , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/virología , Activación de Linfocitos , Linfocitos/inmunología , Linfocitos/metabolismo , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Linfotoxina beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
11.
Proc Natl Acad Sci U S A ; 110(7): 2593-8, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23359703

RESUMEN

Rapid activation of immune responses is necessary for antibacterial defense, but excessive immune activation can result in life-threatening septic shock. Understanding how these processes are balanced may provide novel therapeutic potential in treating inflammatory disease. Fc receptors are crucial for innate immune activation. However, the role of the putative Fc receptor for IgM, known as Toso/Faim3, has to this point been unclear. In this study, we generated Toso-deficient mice and used them to uncover a critical regulatory function of Toso in innate immune activation. Development of innate immune cells was intact in the absence of Toso, but Toso-deficient neutrophils exhibited more reactive oxygen species production and reduced phagocytosis of pathogens compared with controls. Cytokine production was also decreased in Toso(-/-) mice compared with WT animals, rendering them resistant to septic shock induced by lipopolysaccharide. However, Toso(-/-) mice also displayed limited cytokine production after infection with the bacterium Listeria monocytogenes that was correlated with elevated presence of Listeria throughout the body. Accordingly, Toso(-/-) mice succumbed to infections of L. monocytogenes, whereas WT mice successfully eliminated the infection. Taken together, our data reveal Toso to be a unique regulator of innate immune responses during bacterial infection and septic shock.


Asunto(s)
Proteínas Portadoras/inmunología , Granulocitos/inmunología , Inmunidad Innata/inmunología , Listeriosis/inmunología , Activación de Macrófagos/inmunología , Proteínas de la Membrana/inmunología , Monocitos/inmunología , Análisis de Varianza , Animales , Proteínas Portadoras/genética , Cruzamientos Genéticos , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Immunoblotting , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Peroxidasa/metabolismo , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Cell Physiol Biochem ; 36(6): 2379-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26279441

RESUMEN

BACKGROUND: Type I interferon (IFN-I) predisposes to bacterial superinfections, an important problem during viral infection or treatment with interferon-alpha (IFN-α). IFN-I-induced neutropenia is one reason for the impaired bacterial control; however there is evidence that more frequent bacterial infections during IFN-α-treatment occur independently of neutropenia. METHODS: We analyzed in a mouse model, whether Pseudomonas aeruginosa control is influenced by co-infection with the lymphocytic choriomeningitis virus (LCMV). Bacterial titers, numbers of neutrophils and the gene-expression of liver-lysozyme-2 were determined during a 24 hours systemic infection with P. aeruginosa in wild-type and Ifnar(-/-) mice under the influence of LCMV or poly(I:C). RESULTS: Virus-induced IFN-I impaired the control of Pseudomonas aeruginosa. This was associated with neutropenia and loss of lysozyme-2-expression in the liver, which had captured P. aeruginosa. A lower release of IFN-I by poly(I:C)-injection also impaired the bacterial control in the liver and reduced the expression of liver-lysozyme-2. Low concentration of IFN-I after infection with a virulent strain of P. aeruginosa alone impaired the bacterial control and reduced lysozyme-2-expression in the liver as well. CONCLUSION: We found that during systemic infection with P. aeruginosa Kupffer cells quickly controlled the bacteria in cooperation with neutrophils. Upon LCMV-infection this cooperation was disturbed.


Asunto(s)
Interferón Tipo I/farmacología , Virus de la Coriomeningitis Linfocítica/fisiología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/fisiología , Animales , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Inmunidad Innata/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Ratones Endogámicos C57BL , Muramidasa/metabolismo , Neutropenia/inmunología , Neutropenia/patología , Poli I-C/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Bazo/efectos de los fármacos , Bazo/metabolismo , Virulencia/efectos de los fármacos
13.
J Autoimmun ; 62: 11-21, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26094774

RESUMEN

Autoantibodies are a hallmark of autoimmune diseases, such as rheumatoid arthritis, autoimmune hepatitis, and systemic lupus erythematosus (SLE). High titers of anti-nuclear antibodies are used as surrogate marker for SLE, however their contribution to pathogenesis remains unclear. Using murine model of SLE and human samples, we studied the effect of immune stimulation on relapsing of SLE. Although autoantibodies bound to target cells in vivo, only additional activation of CD8(+) T cells converted this silent autoimmunity into overt disease. In mice as well as in humans CD8(+) T cells derived IFN-γ enhanced expression of Fc-receptors on CD11b(+) cells. High expression of Fc-receptors allowed CD11b(+) cells to bind to antibody covered target cells and to destroy them in vivo. We found that autoantibodies induce clinically relevant disease when adaptive immunity, specific for disease non-related antigen, is activated.


Asunto(s)
Antígeno CD11b/metabolismo , Interferón gamma/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Noqueados , Receptores de IgG/genética , Recurrencia
14.
Gut Microbes ; 11(6): 1790-1805, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32521208

RESUMEN

The incidence of gastrointestinal infections continues to increase, and infectious colitis contributes significantly to morbidity and mortality worldwide. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) has been discovered to be strongly involved in the intestinal homeostasis. However, whether intestinal CEACAM1 expression has an impact on the control of infectious colitis remains elusive. Citrobacter rodentium (C. rodentium) is a gram-negative enteric pathogen that induces colonic inflammation in mice, with a critical role for CD4+ T cell but not CD8+ T cell immunity to primary infection. Here, we show that Ceacam1-/- mice are much more susceptible to C. rodentium infection than wildtype mice, which is mediated by a defect in the intestinal barrier and, surprisingly, by a dysregulated CD8+ T cell but not CD4+ T cell response in the colon. CEACAM1 expression is essential for the control of CD8+ T cell immunity, as CEACAM1 deficiency during C. rodentium infection inhibits CD8+ T cell exhaustion. We conclude that CEACAM1 is an important regulator of CD8+ T cell function in the colon, and blocking CEACAM1 signaling to activate CD8+ T cells may have unforeseen side effects.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígeno Carcinoembrionario/inmunología , Citrobacter rodentium/fisiología , Colitis/inmunología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Animales , Linfocitos T CD4-Positivos/inmunología , Antígeno Carcinoembrionario/genética , Colitis/genética , Colitis/microbiología , Colitis/patología , Colon/inmunología , Colon/microbiología , Colon/patología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Front Immunol ; 11: 607889, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584680

RESUMEN

Early and strong production of IFN-I by dendritic cells is important to control vesicular stomatitis virus (VSV), however mechanisms which explain this cell-type specific innate immune activation remain to be defined. Here, using a genome wide association study (GWAS), we identified Integrin alpha-E (Itgae, CD103) as a new regulator of antiviral IFN-I production in a mouse model of vesicular stomatitis virus (VSV) infection. CD103 was specifically expressed by splenic conventional dendritic cells (cDCs) and limited IFN-I production in these cells during VSV infection. Mechanistically, CD103 suppressed AKT phosphorylation and mTOR activation in DCs. Deficiency in CD103 accelerated early IFN-I in cDCs and prevented death in VSV infected animals. In conclusion, CD103 participates in regulation of cDC specific IFN-I induction and thereby influences immune activation after VSV infection.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/virología , Inmunidad Innata , Cadenas alfa de Integrinas/metabolismo , Interferón Tipo I/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/patogenicidad , Animales , Antígenos CD/genética , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno , Cadenas alfa de Integrinas/genética , Ratones de la Cepa 129 , Ratones Endogámicos AKR , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/metabolismo , Vesiculovirus/crecimiento & desarrollo , Replicación Viral
16.
Cell Rep ; 30(11): 3671-3681.e5, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32187540

RESUMEN

Infections can result in a temporarily restricted unresponsiveness of the innate immune response, thereby limiting pathogen control. Mechanisms of such unresponsiveness are well studied in lipopolysaccharide tolerance; however, whether mechanisms of tolerance limit innate immunity during virus infection remains unknown. Here, we find that infection with the highly cytopathic vesicular stomatitis virus (VSV) leads to innate anergy for several days. Innate anergy is associated with induction of apoptotic cells, which activates the Tyro3, Axl, and Mertk (TAM) receptor Mertk and induces high levels of interleukin-10 (IL-10) and transforming growth factor ß (TGF-ß). Lack of Mertk in Mertk-/- mice prevents induction of IL-10 and TGF-ß, resulting in abrogation of innate anergy. Innate anergy is associated with enhanced VSV replication and poor survival after infection. Mechanistically, Mertk signaling upregulates suppressor of cytokine signaling 1 (SOCS1) and SOCS3. Dexamethasone treatment upregulates Mertk and enhances innate anergy in a Mertk-dependent manner. In conclusion, we identify Mertk as one major regulator of innate tolerance during infection with VSV.


Asunto(s)
Anergia Clonal , Inmunidad Innata , Estomatitis Vesicular/enzimología , Estomatitis Vesicular/inmunología , Vesiculovirus/fisiología , Tirosina Quinasa c-Mer/metabolismo , Enfermedad Aguda , Animales , Antivirales/metabolismo , Muerte Celular/efectos de los fármacos , Anergia Clonal/efectos de los fármacos , Dexametasona/farmacología , Activación Enzimática/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Interleucina-10/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Estomatitis Vesicular/virología
17.
Front Immunol ; 11: 1849, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973762

RESUMEN

Immune activation within the tumor microenvironment is one promising approach to induce tumor regression. Certain viruses including oncolytic viruses such as the herpes simplex virus (HSV) and non-oncolytic viruses such as the lymphocytic choriomeningitis virus (LCMV) are potent tools to induce tumor-specific immune activation. However, not all tumor types respond to viro- and/or immunotherapy and mechanisms accounting for such differences remain to be defined. In our current investigation, we used the non-cytopathic LCMV in different human melanoma models and found that melanoma cell lines produced high levels of CCL5 in response to immunotherapy. In vivo, robust CCL5 production in LCMV infected Ma-Mel-86a tumor bearing mice led to recruitment of NK cells and fast tumor regression. Lack of NK cells or CCL5 abolished the anti-tumoral effects of immunotherapy. In conclusion, we identified CCL5 and NK cell-mediated cytotoxicity as new factors influencing melanoma regression during virotherapy.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Quimiocina CCL5/inmunología , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Virus Oncolíticos/inmunología
19.
Front Immunol ; 10: 466, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30930901

RESUMEN

Vesicular stomatitis virus (VSV) is an insect-transmitted rhabdovirus that is neurovirulent in mice. Upon peripheral VSV infection, CD169+ subcapsular sinus (SCS) macrophages capture VSV in the lymph, support viral replication, and prevent CNS neuroinvasion. To date, the precise mechanisms controlling VSV infection in SCS macrophages remain incompletely understood. Here, we show that Toll-like receptor-7 (TLR7), the main sensing receptor for VSV, is central in controlling lymph-borne VSV infection. Following VSV skin infection, TLR7-/- mice display significantly less VSV titers in the draining lymph nodes (dLN) and viral replication is attenuated in SCS macrophages. In contrast to effects of TLR7 in impeding VSV replication in the dLN, TLR7-/- mice present elevated viral load in the brain and spinal cord highlighting their susceptibility to VSV neuroinvasion. By generating novel TLR7 floxed mice, we interrogate the impact of cell-specific TLR7 function in anti-viral immunity after VSV skin infection. Our data suggests that TLR7 signaling in SCS macrophages supports VSV replication in these cells, increasing LN infection and may account for the delayed onset of VSV-induced neurovirulence observed in TLR7-/- mice. Overall, we identify TLR7 as a novel and essential host factor that critically controls anti-viral immunity to VSV. Furthermore, the novel mouse model generated in our study will be of valuable importance to shed light on cell-intrinsic TLR7 biology in future studies.


Asunto(s)
Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Rhabdoviridae/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología , Receptor Toll-Like 7/inmunología , Vesiculovirus/fisiología , Replicación Viral/inmunología , Animales , Encéfalo/inmunología , Encéfalo/virología , Macrófagos/virología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/patología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Médula Espinal/inmunología , Médula Espinal/virología , Receptor Toll-Like 7/genética , Replicación Viral/genética
20.
Nat Commun ; 9(1): 2561, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29967450

RESUMEN

Dysfunction of CD8+ T cells can lead to the development of chronic viral infection. Identifying mechanisms responsible for such T cell dysfunction is therefore of great importance to understand how to prevent persistent viral infection. Here we show using lymphocytic choriomeningitis virus (LCMV) infection that carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is fundamental for recruiting lymphocyte-specific protein kinase (Lck) into the T cell receptor complex to form an efficient immunological synapse. CEACAM1 is essential for activation of CD8+ T cells, and the absence of CEACAM1 on virus-specific CD8+ T cells limits the antiviral CD8+ T cell response. Treatment with anti-CEACAM1 antibody stabilizes Lck in the immunological synapse, prevents CD8+ T cell exhaustion, and improves control of virus infection in vivo. Treatment of human virus-specific CD8+ T cells with anti-CEACAM1 antibody similarly enhances their proliferation. We conclude that CEACAM1 is an important regulator of virus-specific CD8+ T cell functions in mice and humans and represents a promising therapeutic target for modulating CD8+ T cells.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD8-positivos/inmunología , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Traslado Adoptivo , Animales , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/metabolismo , Antígeno Carcinoembrionario/genética , Quimera , Enfermedad Crónica , Femenino , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA