Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
3.
Clin Exp Immunol ; 203(3): 448-457, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33040328

RESUMEN

Severe combined immunodeficiency (SCID) is fatal if not treated with immune reconstitution. In Egypt, T- B+ SCID accounts for 38·5% of SCID diagnoses. An accurate genetic diagnosis is essential for choosing appropriate treatment modalities and for offering genetic counseling to the patient's family. The objectives of this study were to describe the clinical, immunological and molecular characteristics of a cohort of twenty Egyptian patients with T- B+ SCID. The initial diagnosis (based on clinical features and flow cytometry) was followed by molecular investigation (whole-exome sequencing). All patients had the classic clinical picture for SCID, including failure to thrive (n = 20), oral candidiasis (n = 17), persistent diarrhea (n = 14), pneumonia (n = 13), napkin dermatitis (n = 10), skin rash (n = 7), otitis media (n = 3) and meningitis (n = 2). The onset of manifestations was at the age of 2·4 ± 1·6 months and diagnosis at the age of 6·7 ± ·5 months, giving a diagnostic delay of 4·3 months. JAK3 gene variants were most frequent (n = 12) with three novel variants identified, followed by IL2Rγ variants (n = 6) with two novel variants. IL7Rα and CD3ε variants were found once, with a novel variant each. T- B+ NK- SCID accounted for approximately 90% of the Egyptian patients with T- B+ SCID. Of these T- B+ NK- SCID cases, 60% were autosomal recessive syndromes caused by JAK3 mutations and 30% were X-linked syndromes. It might be useful to sequence the JAK3 gene (i.e. targeted Sanger sequencing) in all T- B+ SCID patients, especially after X-linked SCID has been ruled out. Hence, no more than 10% of T- B+ SCID patients might require next-generation for a molecular diagnosis.


Asunto(s)
Secuenciación del Exoma/métodos , Janus Quinasa 3/genética , Mutación , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Consanguinidad , Egipto , Salud de la Familia , Femenino , Humanos , Lactante , Recién Nacido , Subunidad gamma Común de Receptores de Interleucina/genética , Janus Quinasa 3/deficiencia , Recuento de Linfocitos , Masculino , Linaje , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/metabolismo
4.
Front Immunol ; 11: 616898, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584707

RESUMEN

The antigen-independent, strong proliferative responses of naive CD8+ T cells have been well demonstrated in a particular strain of mice lacking IL-2 receptors. This type of proliferation is mainly driven by common gamma-chain (γc) cytokines, such as IL-2, IL-7, and IL-15, present at abnormally high levels in these mice. Similarly, in the present study, we showed that mice lacking Janus kinase 3 (Jak3), a tyrosine kinase crucial for γc cytokine signaling, could induce strong proliferation of adoptively transferred naive CD8+ T cells. This proliferation was also independent of antigenic stimulation, but heavily dependent on IL-2, as evidenced by the failure of proliferation of adoptively transferred IL-2 receptor alpha- and beta-chain-deficient naive CD8+ T cells. Consistent with this, Jak3-/- mice showed elevated serum levels of IL-2 compared to wild-type mice, and interestingly, IL-2 production was due to high levels of accumulation of activated CD4+ T cells in Jak3-/- mice along with defective CD4+ T regulatory cells. Collectively, these findings reveal previously unidentified unique immune contexts of Jak3-/- mice that cause robust IL-2-driven T cell expansion and have a clinical implication for designing a treatment strategy for human patients with loss-of-function genetic mutations of Jak3.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-2/inmunología , Janus Quinasa 3/deficiencia , Janus Quinasa 3/inmunología , Activación de Linfocitos/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Antiviral Res ; 149: 78-88, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28893602

RESUMEN

Employing NOD/SCID/Jak3-/- mice transplanted with human PBMCs (hNOJ mice) and replication-competent, red-fluorescent-protein (mCherry; mC)-labeled HIV-1JR-FL (HIVmC), we examined whether early antiretroviral treatment blocked the establishment of HIV-1 infection. The use of hNOJ mice and HIVmC enabled us to visually locate infection foci and to examine the early dynamics of HIVmC infection without using a large amount of antiretroviral unlike in non-human primate models. Although when raltegravir (RAL) administration was begun 1 day after intraperitoneal (ip) inoculation of HIVmC, no plasma p24 or plasma HIV-1-RNA (pRNA) were detected in 10 of 12 hNOJ (hNOJmCRAL+) mice as assessed on the last day of the 14-day continuous twice-daily RAL administration, all 10 untreated hNOJmC (hNOJmCRAL-) mice became positive for p24 and pRNA and had significantly swollen lymph nodes in peritoneal cavity and abundant p24+/mC+/CD3+/CD4+ T cells and p24+/mC+/CD68+ monocytes/macrophages were identified in their omenta and mesenteric lymphoid tissues/lymph nodes upon necropsy of the mice on day 14. In 12 hNOJmCRAL+ mice, no significantly swollen lymph nodes were seen compared to hNOJmCRAL- mice; however, in the omentum of the 2 hNOJmCRAL+ mice that were positive for pRNA and in site RNA, mC+/p24+/CD3+/CD83+ cells were identified, suggesting that viral breakthrough occurred later in the observation period. The present data suggest that the use of hNOJ mouse model and HIVmC may shed light on the study of early-phase dynamics of HIV-1 infection and cellular events in post-exposure/pre-exposure prophylaxis.


Asunto(s)
Fármacos Anti-VIH/farmacología , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Profilaxis Posexposición , Raltegravir Potásico/farmacología , Animales , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Vectores Genéticos/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/prevención & control , VIH-1/genética , Humanos , Inmunohistoquímica , Janus Quinasa 3/deficiencia , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Profilaxis Posexposición/métodos , Carga Viral , Proteína Fluorescente Roja
6.
Medicine (Baltimore) ; 96(42): e7989, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29049190

RESUMEN

RATIONALE: With the progress of sequencing technology, an increasing number of atypical primary immunodeficiency (PID) patients have been discovered, including Janus kinase 3 (JAK3) gene deficiency. PATIENT CONCERNS: We report a patient who presented with chronic active Epstein-Barr virus (CAEBV) infection but responded poorly to treatment with ganciclovir. DIAGNOSES: Next-generation sequencing (NGS) was performed, including all known PID genes, after which Sanger sequencing was performed to verify the results. Genetic analysis revealed that our patient had 2 novel compound heterozygous mutations of JAK3, a gene previously reported to cause a rare form of autosomal recessive severe combined immunodeficiency with recurrent infections. The p.H27Q mutation came from his father, while p. R222H from his mother. Thus, his diagnosis was corrected for JAK3-deficiency PID and CAEBV. INTERVENTIONS: Maintenance treatment of subcutaneous injection of recombinant human interferon α-2a was given to our patient with 2 MU, 3 times a week. OUTCOMES: Interferon alpha was applied and the EBV infection was gradually controlled and his symptoms ameliorated remarkably. Our patient is in good health now and did not have relapses. LESSONS: The diagnoses of PID should be taken into consideration when CAEBV patients respond poorly to conventional treatments. Good results of our patient indicate that interferon α-2a may be an alternative treatment for those who are unwilling to accept hematopoietic stem cell transplantation (HSCT) like our patient. Literature review identified 59 additional cases of JAK3 deficiency with various infections.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4 , Síndromes de Inmunodeficiencia/genética , Janus Quinasa 3/deficiencia , Antivirales/uso terapéutico , Niño , Enfermedad Crónica , Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Infecciones por Virus de Epstein-Barr/virología , Ganciclovir/uso terapéutico , Herpesvirus Humano 4/efectos de los fármacos , Humanos , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Síndromes de Inmunodeficiencia/virología , Interferón alfa-2 , Interferón-alfa/administración & dosificación , Masculino , Proteínas Recombinantes/administración & dosificación , Resultado del Tratamiento
7.
Antiviral Res ; 144: 83-92, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28392419

RESUMEN

We attempted to elucidate early-phase dynamics of HIV-1 infection using replication-competent, red-fluorescent-protein (mCherry)-labeled HIV-1JR-FL (HIVJR-FLmC) and NOD/SCID/Jak3-/- mice transplanted with Individual-A's human peripheral blood mononuclear cells (hPBMC)(hNOJ mice). On day 7 following HIVJR-FLmC inoculation, mCherry-signal-emitting infection foci were readily identified in the subserosa of 10 of 10 HIVJR-FLmC-inoculated hNOJ mice, although infection foci were not located without the mCherry signal in unlabeled HIV-1JR-FL-inoculated mice (n = 6). Even on day 14, infection foci were hardly located in the unlabeled HIV-1JR-FL-inoculated mice, while in all of 7 HIVJR-FLmC-inoculated hNOJ mice examined, mCherry-signal-emitting lymph nodes were easily identified, in which active viral replication was present. On day 14, a significantly larger number of mesenteric lymph nodes were seen in HIVJR-FLmC-exposed hNOJ mice than in HIVJR-FLmC-unexposed mice (P = 0.0025). The weights of mesenteric lymph nodes of those HIVJR-FLmC-exposed hNOJ mice were also greater than those of HIVJR-FLmC-unexposed mice (P = 0.0005). When hNOJ mice were inoculated with HIVJR-FLmC-exposed hPBMC from Individual-B, significantly greater viremia was seen than in cell-free HIVJR-FLmC-inoculated hNOJ mice as examined on day 7. In the lymph nodes of those mice inoculated with HIVJR-FLmC-exposed hPBMC from Individual-B, a substantial number of Individual-B's HIVJR-FLmC-infected cells were identified together with Individual-A's cells as examined on day 14. The present HIVJR-FLmC-infected mouse model represents the first system reported using traceable HIVJR-FLmC and human target cells, not using SIV or simian cells, which should be of utility in studies of early-phases of HIV-1 transmission and in evaluating the effects of potential agents for post-exposure and pre-exposure prophylaxis.


Asunto(s)
Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , Leucocitos Mononucleares/virología , Proteínas Luminiscentes/análisis , Animales , Modelos Animales de Enfermedad , Genes Reporteros , VIH-1/genética , Janus Quinasa 3/deficiencia , Proteínas Luminiscentes/genética , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Coloración y Etiquetado , Proteína Fluorescente Roja
9.
Nat Immunol ; 17(11): 1291-1299, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27618553

RESUMEN

Innate lymphoid cells (ILCs) have potent immunological functions in experimental conditions in mice, but their contributions to immunity in natural conditions in humans have remained unclear. We investigated the presence of ILCs in a cohort of patients with severe combined immunodeficiency (SCID). All ILC subsets were absent in patients with SCID who had mutation of the gene encoding the common γ-chain cytokine receptor subunit IL-2Rγ or the gene encoding the tyrosine kinase JAK3. T cell reconstitution was observed in patients with SCID after hematopoietic stem cell transplantation (HSCT), but the patients still had considerably fewer ILCs in the absence of myeloablation than did healthy control subjects, with the exception of rare cases of reconstitution of the ILC1 subset of ILCs. Notably, the ILC deficiencies observed were not associated with any particular susceptibility to disease, with follow-up extending from 7 years to 39 years after HSCT. We thus report here selective ILC deficiency in humans and show that ILCs might be dispensable in natural conditions, if T cells are present and B cell function is preserved.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Adolescente , Adulto , Animales , Biomarcadores , Niño , Modelos Animales de Enfermedad , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Janus Quinasa 3/deficiencia , Recuento de Linfocitos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Linfocitos/metabolismo , Linfopenia/sangre , Linfopenia/etiología , Ratones , Ratones Noqueados , Fenotipo , Inmunodeficiencia Combinada Grave/sangre , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/metabolismo , Inmunodeficiencia Combinada Grave/terapia , Piel/inmunología , Piel/patología
10.
J Pediatr ; 173: 235-7, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26996725

RESUMEN

A 35-day-old female with severe combined immunodeficiency developed cytomegalovirus (CMV) meningitis before undergoing hematopoietic stem cell transplantation. Strategies for timely diagnosis of neonates with congenital or acquired CMV infection and prevention of CMV acquisition in the era of universal newborn severe combined immunodeficiency screening are needed.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Meningitis Viral/complicaciones , Inmunodeficiencia Combinada Grave/complicaciones , Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Recién Nacido , Janus Quinasa 3/deficiencia , Meningitis Viral/diagnóstico , Meningitis Viral/tratamiento farmacológico , Inmunodeficiencia Combinada Grave/terapia
11.
Cell Physiol Biochem ; 36(6): 2287-98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26279433

RESUMEN

BACKGROUND/AIMS: Janus kinase 3 (JAK3), a tyrosine kinase mainly expressed in hematopoietic cells, participates in the signaling stimulating cell proliferation. The kinase is expressed in dendritic cells (DCs), antigen presenting cells involved in the initiation and regulation of antigen-specific T-cell responses. Dendritic cell function is regulated by cytosolic Ca(2+) activity ([Ca(2+)]i). Mediators increasing [Ca(2+)]i in DCs include ATP and the chemokine receptor CXCR4 ligand CXCL12. The present study explored, whether JAK3 participates in the regulation of [Ca(2+)]i in DCs. METHODS: Fura-2 fluorescence was employed to determine [Ca(2+)]i, and whole cell patch clamp to decipher electrogenic transport in immature DCs isolated from bone marrow of JAK3-knockout (jak3(-/-)) or wild-type mice (jak3(+/+)). RESULTS: Without treatment, [Ca(2+)]i was similar in jak3(-/-) and jak3(+/+) DCs. Addition of ATP (100 µM) was followed by transient increase of [Ca(2+)]i reflecting Ca(2+) release from intracellular stores, an effect significantly less pronounced in jak3(-/-) DCs than in jak3(+/+) DCs. CXCL12 administration was followed by a sustained increase of [Ca(2+)]i reflecting receptor operated Ca(2+) entry, an effect significantly less rapid in jak3(-/-) DCs than in jak3(+/+) DCs. In addition, the Ca(2+) release-activated Ca(2+) channel (CRAC) current triggered by IP3-induced Ca(2+) store depletion and CXCL12 was significantly higher in DCs from jak3(+/+) mice than in jak3(-/-) mice. Inhibition of sarcoendoplasmatic reticulum Ca(2+)-ATPase (SERCA) by thapsigargin (1 µM) in the absence of extracellular Ca(2+) was followed by a transient increase of [Ca(2+)]i reflecting Ca(2+) release from intracellular stores, and subsequent readdition of extracellular Ca(2+) in the continued presence of thapsigargin was followed by a sustained increase of [Ca(2+)]i reflecting store operated Ca(2+) entry (SOCE). Both, Ca(2+) release from intracellular stores and SOCE were again significantly lower in jak3(-/-) DCs than in jak3(+/+) DCs. Pretreatment of jak3(+/+) DCs with JAK inhibitor WHI-P154 (22 µM, 10 minutes or 24 hours) significantly blunted both thapsigargin induced Ca(2+) release and subsequent SOCE. Abrupt replacement of Na(+) containing (130 mM) and Ca(2+) free (0 mM) extracellular bath by Na(+) free (0 mM) and Ca(2+) containing (2 mM) extracellular bath increased [Ca(2+)]i reflecting Na(+)/Ca(2+) exchanger activity, an effect again significantly less pronounced in jak3(-/-) DCs than in jak3(+/+) DCs. CONCLUSIONS: JAK3 deficiency is followed by down-regulation of cytosolic Ca(2+) release, receptor and store operated Ca(2+) entry and Na(+)/Ca(2+) exchanger activity in DCs.


Asunto(s)
Calcio/metabolismo , Células Dendríticas/metabolismo , Janus Quinasa 3/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Adenosina Trifosfato/farmacología , Animales , Quimiocina CXCL12/farmacología , Células Dendríticas/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Janus Quinasa 3/deficiencia , Masculino , Ratones , Quinazolinas/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Tapsigargina/farmacología
12.
Kidney Int ; 87(4): 728-37, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25493954

RESUMEN

Calcitriol, a powerful regulator of phosphate metabolism and immune response, is generated by 25-hydroxyvitamin D 1α-hydroxylase in the kidney and macrophages. Renal 1α-hydroxylase expression is suppressed by Klotho and FGF23, the expression of which is stimulated by calcitriol. Interferon γ (INFγ) regulates 1α-hydroxylase expression in macrophages through transcription factor interferon regulatory factor-1. INFγ-signaling includes Janus kinase 3 (JAK3) but a role of JAK3 in the regulation of 1α-hydroxylase expression and mineral metabolism has not been shown. Thus, the impact of JAK3 deficiency on calcitriol formation and phosphate metabolism was measured. Renal interferon regulatory factor-1 and 1α-hydroxylase transcript levels, serum calcitriol and FGF23 levels, intestinal phosphate absorption as well as absolute and fractional renal phosphate excretion were significantly higher in jak3 knockout than in wild-type mice. Coexpression of JAK3 increased the phosphate-induced current in renal sodium-phosphate cotransporter-expressing Xenopus oocytes. Thus, JAK3 is a powerful regulator of 1α-hydroxylase expression and phosphate transport. Its deficiency leads to marked derangement of phosphate metabolism.


Asunto(s)
25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , Calcitriol/sangre , Janus Quinasa 3/metabolismo , Riñón/enzimología , Fosfatos/metabolismo , ARN Mensajero/metabolismo , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/análisis , Animales , Calbindinas/genética , Calcitriol/biosíntesis , Heces/química , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Factor 1 Regulador del Interferón/análisis , Factor 1 Regulador del Interferón/genética , Mucosa Intestinal/metabolismo , Janus Quinasa 3/deficiencia , Janus Quinasa 3/genética , Riñón/química , Masculino , Ratones , Ratones Noqueados , Oocitos/enzimología , Fosfatos/análisis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Regulación hacia Arriba , Xenopus
14.
PLoS One ; 9(8): e106056, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25170898

RESUMEN

BACKGROUND: Up-regulation and association of nuclear factor kappa B (NF-κB) with carcinogenesis and tumor progression has been reported in several malignancies. In the current study, expression of NF-κB in cholangiocarcinoma (CCA) patient tissues and its clinical significance were determined. The possibility of using NF-κB as the therapeutic target of CCA was demonstrated. METHODOLOGY: Expression of NF-κB in CCA patient tissues was determined using immunohistochemistry. Dehydroxymethylepoxyquinomicin (DHMEQ), a specific NF-κB inhibitor, was used to inhibit NF-κB action. Cell growth was determined using an MTT assay, and cell apoptosis was shown by DNA fragmentation, flow cytometry and immunocytofluorescent staining. Effects of DHMEQ on growth and apoptosis were demonstrated in CCA cell lines and CCA-inoculated mice. DHMEQ-induced apoptosis in patient tissues using a histoculture drug response assay was quantified by TUNEL assay. PRINCIPAL FINDINGS: Normal bile duct epithelia rarely expressed NF-κB (subunits p50, p52 and p65), whereas all CCA patient tissues (n  =  48) over-expressed all NF-κB subunits. Inhibiting NF-κB action by DHMEQ significantly inhibited growth of human CCA cell lines in a dose- and time-dependent manner. DHMEQ increased cell apoptosis by decreasing the anti-apoptotic protein expressions-Bcl-2, XIAP-and activating caspase pathway. DHMEQ effectively reduced tumor size in CCA-inoculated mice and induced cell apoptosis in primary histocultures of CCA patient tissues. CONCLUSIONS: NF-κB was over-expressed in CCA tissues. Inhibition of NF-κB action significantly reduced cell growth and enhanced cell apoptosis. This study highlights NF-κB as a molecular target for CCA therapy.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos , Colangiocarcinoma/metabolismo , Hígado/metabolismo , FN-kappa B/biosíntesis , Opistorquiasis/metabolismo , Animales , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Neoplasias de los Conductos Biliares/complicaciones , Neoplasias de los Conductos Biliares/patología , Western Blotting , Colangiocarcinoma/complicaciones , Colangiocarcinoma/patología , Ciclohexanonas/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Interacciones Huésped-Parásitos/efectos de los fármacos , Humanos , Inmunohistoquímica , Janus Quinasa 3/deficiencia , Janus Quinasa 3/genética , Hígado/parasitología , Hígado/patología , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , FN-kappa B/antagonistas & inhibidores , Opistorquiasis/complicaciones , Opistorquiasis/parasitología , Opisthorchis/fisiología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Eur J Cancer ; 50(10): 1836-1846, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24726056

RESUMEN

BACKGROUND: Recently, the critical role of CD47 on the surface of resistant cancer cells has been proposed in their evasion of immunosurveillance. Primary effusion lymphoma (PEL) is a subtype of aggressive non-Hodgkin lymphoma that shows serous lymphomatous effusion in body cavities, especially in advanced acquired immunodeficiency syndrome (AIDS). PEL is resistant to conventional chemotherapy and has a poor prognosis. In this study, we evaluated the effect of anti-CD47 antibody (Ab) on PEL in vitro and in vivo. METHODS: Surface CD47 of PEL cell lines was examined by flow cytometry. Efficacy of knocking down CD47 or anti-CD47 Ab-mediated phagocytosis against PEL was evaluated using mouse peritoneal macrophages and human macrophages in vitro. Primary PEL cells were injected intraperitoneally into NOD/Rag-2/Jak3 double-deficient (NRJ) mice to establish a direct xenograft mouse model. RESULTS: Surface CD47 of PEL cell lines was highly expressed. Knocking down CD47 and anti-CD47 Ab promoted phagocytic activities of macrophages in a CD47 expression-dependent manner in vitro. Treatment with anti-CD47 Ab inhibited ascite formation and organ invasion completely in vivo compared with control IgG-treated mice. CONCLUSION: CD47 plays the pivotal role in the immune evasion of PEL cells in body cavities. Therapeutic antibody targeting of CD47 could be an effective therapy for PEL.


Asunto(s)
Anticuerpos/farmacología , Antineoplásicos/farmacología , Antígeno CD47/inmunología , Linfoma de Efusión Primaria/tratamiento farmacológico , Macrófagos Peritoneales/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Escape del Tumor/efectos de los fármacos , Adulto , Animales , Antígeno CD47/metabolismo , Línea Celular , Técnicas de Cocultivo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Humanos , Janus Quinasa 3/deficiencia , Janus Quinasa 3/genética , Linfoma de Efusión Primaria/genética , Linfoma de Efusión Primaria/inmunología , Linfoma de Efusión Primaria/metabolismo , Linfoma de Efusión Primaria/patología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Invasividad Neoplásica , Interferencia de ARN , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Kidney Blood Press Res ; 37(4-5): 514-20, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24281140

RESUMEN

BACKGROUND/AIMS: The Janus kinase 3 JAK3 participates in the signaling of immune cells. Lack of JAK3 triggers inflammatory bowel disease, which in turn has been shown to affect intestinal activity of the epithelial Na(+) channel ENaC and thus colonic sodium absorption. At least in theory, inflammatory bowel disease in JAK3-deficient mice could lead to intestinal salt loss compromizing extracellular volume maintenance and blood pressure regulation. The present study thus explored whether JAK3 deficiency impacts on colonic ENaC activity, fecal Na(+) exretion, blood pressure and extracellular fluid volume regulation. METHODS: Experiments were performed in gene-targeted mice lacking functional JAK3 (jak3(-/-)) and in wild type mice (jak3(+/+)). Colonic ENaC activity was estimated from amiloride-sensitive current in Ussing chamber experiments, fecal, serum and urinary Na(+) concentration by flame photometry, blood pressure by the tail cuff method and serum aldosterone levels by immunoassay. RESULTS: The amiloride (50 µM)-induced deflection of the transepithelial potential difference was significantly lower and fecal Na(+) excretion significantly higher in jak3(-/-) mice than in jak3(+/+) mice. Moreover, systolic arterial blood pressure was significantly lower and serum aldosterone concentration significantly higher in jak3(-/-) mice than in jak3(+/+) mice. Both, absolute and fractional renal Na(+) excretion were significantly lower in jak3(-/-) mice than in jak3(+/+) mice. CONCLUSIONS: JAK3 deficiency leads to impairment of colonic ENaC activity with intestinal Na(+) loss, decrease of blood pressure, increased aldosterone release and subsequent stimulation of renal tubular Na(+) reabsorption.


Asunto(s)
Colon/metabolismo , Canales Epiteliales de Sodio/metabolismo , Motilidad Gastrointestinal/fisiología , Hipovolemia/metabolismo , Janus Quinasa 3/deficiencia , Sodio/metabolismo , Animales , Femenino , Hipovolemia/diagnóstico , Masculino , Ratones , Ratones Noqueados
18.
PLoS One ; 7(8): e42776, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22880104

RESUMEN

Humanized mice are expected to be useful as small animal models for in vivo studies on the pathogenesis of infectious diseases. However, it is well known that human CD8(+) T cells cannot differentiate into effector cells in immunodeficient mice transplanted with only human CD34(+) hematopoietic stem cells (HSCs), because human T cells are not educated by HLA in the mouse thymus. We here established HLA-B*51:01 transgenic humanized mice by transplanting human CD34(+) HSCs into HLA-B*51:01 transgenic NOD/SCID/Jak3(-/-) mice (hNOK/B51Tg mice) and investigated whether human effector CD8(+) T cells would be elicited in the mice or in those infected with HIV-1 NL4-3. There were no differences in the frequency of late effector memory and effector subsets (CD27(low)CD28(-)CD45RA(+/-)CCR7(-) and CD27(-)CD28(-)CD45RA(+/-)CCR7(-), respectively) among human CD8(+) T cells and in that of human CD8(+) T cells expressing CX3CR1 and/or CXCR1 between hNOK/B51Tg and hNOK mice. In contrast, the frequency of late effector memory and effector CD8(+) T cell subsets and of those expressing CX3CR1 and/or CXCR1 was significantly higher in HIV-1-infected hNOK/B51Tg mice than in uninfected ones, whereas there was no difference in that of these subsets between HIV-1-infected and uninfected hNOK mice. These results suggest that hNOK/B51Tg mice had CD8(+) T cells that were capable of differentiating into effector T cells after viral antigen stimulation and had a greater ability to elicit effector CD8(+) T cells than hNOK ones.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Antígeno HLA-B51/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Humanos , Janus Quinasa 3/deficiencia , Janus Quinasa 3/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fenotipo , Receptores de Quimiocina/inmunología , Receptores de Interleucina-8A/inmunología
19.
Pediatr Infect Dis J ; 31(2): 204-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22138680

RESUMEN

Disseminated cryptococcal infection is the second most common cause of death after tuberculosis in acquired immune deficiency syndrome patients. Surprisingly, it has been reported only in few patients with primary immunodeficiency diseases. Herein, we report the clinical presentation and outcome of a 23-month-old boy with novel JAK3 mutation severe combined immunodeficiency disease complicated by severe disseminated cryptococcal infection.


Asunto(s)
Criptococosis/diagnóstico , Criptococosis/patología , Janus Quinasa 3/deficiencia , Inmunodeficiencia Combinada Grave/complicaciones , Trasplante de Células Madre , Humanos , Lactante , Janus Quinasa 3/genética , Masculino , Inmunodeficiencia Combinada Grave/genética , Resultado del Tratamiento
20.
Cell Physiol Biochem ; 27(5): 547-56, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691072

RESUMEN

Janus kinase 3, a tyrosine kinase expressed in haematopoetic tissues, plays a decisive role in T-lymphocyte survival. JAK3 deficiency leads to (Severe) Combined Immunodeficiency (SCID) resulting from enhanced lymphocyte apoptosis. JAK3 is activated by phosphorylation. Nothing is known about expression of JAK3 in erythrocytes, which may undergo apoptosis-like cell death (eryptosis) characterized by cell membrane scrambling with phosphatidylserine exposure and cell shrinkage. Triggers of eryptosis include energy depletion. The present study utilized immunohistochemistry and confocal microscopy to test for JAK3 expression and phosphorylation, and FACS analysis to determine phosphatidylserine exposure (annexin binding) and cell volume (forward scatter). As a result, JAK3 was expressed in erythrocytes and phosphorylated following 24h and 48h glucose depletion. Forward scatter was slightly but significantly smaller in erythrocytes from JAK3-deficient mice (jak3(-/-)) than in erythrocytes from wild type mice (jak3(+/+)). Annexin V binding was similarly low in both genotypes. The JAK3 inhibitors WHI-P131/JANEX-1 (4-(4'-Hydroxyphenyl)amino-6,7-dimethoxyquinazoline, 156µM) and WHI-P154 (4-[(3'-Bromo-4'-hydroxyphenyl)amino]-6,7-dimethoxyquinazoline, 11.2µM) did not significantly modify annexin V binding or forward scatter. Glucose depletion increased annexin V binding, an effect significantly blunted in jak3(-/-) erythrocytes and in the presence of the JAK3 inhibitors. The observations disclose a completely novel role of Janus kinase 3, i.e. the triggering of cell membrane scrambling in energy depleted erythrocytes.


Asunto(s)
Eritrocitos/enzimología , Glucosa/farmacología , Janus Quinasa 3 , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/análisis , Adenosina Trifosfato/metabolismo , Animales , Anexina A5/análisis , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Tamaño de la Célula , Recuento de Eritrocitos , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Femenino , Citometría de Flujo , Eliminación de Gen , Glucosa/deficiencia , Humanos , Inmunohistoquímica , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/biosíntesis , Janus Quinasa 3/deficiencia , Janus Quinasa 3/genética , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Fosfatidilserinas/análisis , Fosfatidilserinas/metabolismo , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA