Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Environ Sci Technol ; 58(24): 10706-10716, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38850513

RESUMEN

Most previous measurements of oxidized mercury were collected using a method now known to be biased low. In this study, a dual-channel system with an oxidized mercury detection limit of 6-12 pg m-3 was deployed alongside a permeation tube-based automated calibrator at a mountain top site in Steamboat Springs Colorado, USA, in 2021 and 2022. Permeation tubes containing elemental mercury and mercury halides were characterized via an International System of Units (SI)-traceable gravimetric method and gas chromatography/mass spectrometry before deployment in the calibrator. The dual-channel system recovered 97 ± 4 and 100 ± 8% (±standard deviation) of injected elemental mercury and HgBr2, respectively. Total Hg permeation rates and Hg speciation from the gravimetric method, the chromatography system, the dual-channel system, and an independent SI-traceable measurement method performed at the Jozef Stefan Institute laboratory were all comparable within the respective uncertainties of each method. These are the first measurements of oxidized mercury at low environmental concentrations that have been verified against an SI-traceable calibration system in field conditions while sampling ambient air, and they show that accurate, routinely calibrated oxidized mercury measurements are achievable.


Asunto(s)
Monitoreo del Ambiente , Mercurio , Oxidación-Reducción , Mercurio/análisis , Calibración , Monitoreo del Ambiente/métodos , Atmósfera/química , Contaminantes Atmosféricos/análisis , Colorado , Cromatografía de Gases y Espectrometría de Masas
2.
Malar J ; 17(1): 114, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540197

RESUMEN

BACKGROUND: Human-immune-system humanized mouse models can bridge the gap between humans and conventional mice for testing human vaccines. The HLA-expressing humanized DRAGA (HLA-A2.HLA-DR4.Rag1KO.IL2RγcKO.NOD) mice reconstitute a functional human-immune-system and sustain the complete life cycle of Plasmodium falciparum. Herein, the DRAGA mice were investigated for immune responses following immunization with live P. falciparum sporozoites under chloroquine chemoprophylaxis (CPS-CQ), an immunization approach that showed in human trials to confer pre-erythrocytic immunity. RESULTS: The CPS-CQ immunized DRAGA mice (i) elicited human CD4 and CD8 T cell responses to antigens expressed by P. falciparum sporozoites (Pfspz) and by the infected-red blood cells (iRBC). The Pfspz-specific human T cell responses were found to be systemic (spleen and liver), whereas the iRBCs-specific human T cell responses were more localized to the liver, (ii) elicited stronger antibody responses to the Pfspz than to the iRBCs, and (iii) they were protected against challenge with infectious Pfspz but not against challenge with iRBCs. CONCLUSIONS: The DRAGA mice represent a new pre-clinical model to investigate the immunogenicity and protective efficacy of P. falciparum malaria vaccine candidates.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Cloroquina/uso terapéutico , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Animales , Formación de Anticuerpos , Antimaláricos/uso terapéutico , Humanos , Ratones , Ratones Transgénicos
3.
Malar J ; 13: 386, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25266106

RESUMEN

BACKGROUND: Malaria is a deadly infectious disease affecting millions of people in tropical and sub-tropical countries. Among the five species of Plasmodium parasites that infect humans, Plasmodium falciparum accounts for the highest morbidity and mortality associated with malaria. Since humans are the only natural hosts for P. falciparum, the lack of convenient animal models has hindered the understanding of disease pathogenesis and prompted the need of testing anti-malarial drugs and vaccines directly in human trials. Humanized mice hosting human cells represent new pre-clinical models for infectious diseases that affect only humans. In this study, the ability of human-immune-system humanized HLA-DR4.RagKO.IL2RγcKO.NOD (DRAG) mice to sustain infection with P. falciparum was explored. METHODS: Four week-old DRAG mice were infused with HLA-matched human haematopoietic stem cells (HSC) and examined for reconstitution of human liver cells and erythrocytes. Upon challenge with infectious P. falciparum sporozoites (NF54 strain) humanized DRAG mice were examined for liver stage infection, blood stage infection, and transmission to Anopheles stephensi mosquitoes. RESULTS: Humanized DRAG mice reconstituted human hepatocytes, Kupffer cells, liver endothelial cells, and erythrocytes. Upon intravenous challenge with P. falciparum sporozoites, DRAG mice sustained liver to blood stage infection (average 3-5 parasites/microlitre blood) and allowed transmission to An. stephensi mosquitoes. Infected DRAG mice elicited antibody and cellular responses to the blood stage parasites and self-cured the infection by day 45 post-challenge. CONCLUSIONS: DRAG mice represent the first human-immune-system humanized mouse model that sustains the complex vertebrate life cycle of P. falciparum without the need of exogenous injection of human hepatocytes/erythrocytes or P. falciparum parasite adaptation. The ability of DRAG mice to elicit specific human immune responses to P. falciparum parasites may help deciphering immune correlates of protection and to identify protective malaria antigens.


Asunto(s)
Malaria Falciparum/parasitología , Ratones Transgénicos/parasitología , Animales , Anopheles/parasitología , Anticuerpos Antiprotozoarios/sangre , Eritrocitos/citología , Femenino , Hepatocitos/citología , Humanos , Macrófagos del Hígado/citología , Malaria Falciparum/inmunología , Ratones , Ratones Transgénicos/inmunología , Parasitemia/inmunología , Parasitemia/parasitología , Plasmodium falciparum/inmunología , Esporozoítos/inmunología
4.
Front Cell Infect Microbiol ; 14: 1367566, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38983114

RESUMEN

Humanized mouse models are valuable tools for investigating the human immune system in response to infection and injury. We have previously described the human immune system (HIS)-DRAGA mice (HLA-A2.HLA-DR4.Rag1KO.IL-2RgKO.NOD) generated by infusion of Human Leukocyte Antigen (HLA)-matched, human hematopoietic stem cells from umbilical cord blood. By reconstituting human cells, the HIS-DRAGA mouse model has been utilized as a "surrogate in vivo human model" for infectious diseases such as Human Immunodeficiency Virus (HIV), Influenza, Coronavirus Disease 2019 (COVID-19), scrub typhus, and malaria. This humanized mouse model bypasses ethical concerns about the use of fetal tissues for the humanization of laboratory animals. Here in, we demonstrate the presence of human microglia and T cells in the brain of HIS-DRAGA mice. Microglia are brain-resident macrophages that play pivotal roles against pathogens and cerebral damage, whereas the brain-resident T cells provide surveillance and defense against infections. Our findings suggest that the HIS-DRAGA mouse model offers unique advantages for studying the functions of human microglia and T cells in the brain during infections, degenerative disorders, tumors, and trauma, as well as for testing therapeutics in these pathological conditions.


Asunto(s)
Encéfalo , Modelos Animales de Enfermedad , Microglía , Linfocitos T , Animales , Microglía/inmunología , Humanos , Ratones , Encéfalo/inmunología , Linfocitos T/inmunología , COVID-19/inmunología , SARS-CoV-2/inmunología
5.
Hum Vaccin Immunother ; 18(5): 2048622, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-35348437

RESUMEN

We report a Human Immune System (HIS)-humanized mouse model ("DRAGA": HLA-A2.HLA-DR4.Rag1KO.IL-2 RγcKO.NOD) for COVID-19 research. DRAGA mice express transgenically HLA-class I and class-II molecules in the mouse thymus to promote human T cell development and human B cell Ig-class switching. When infused with human hematopoietic stem cells from cord blood reconstitute a functional human immune system, as well as human epi/endothelial cells in lung and upper respiratory airways expressing the human ACE2 receptor for SARS-CoV-2. The DRAGA mice were able to sustain SARS-CoV-2 infection for at least 25 days. Infected mice showed replicating virus in the lungs, deteriorating clinical condition, and human-like lung immunopathology including human lymphocyte infiltrates, microthrombi and pulmonary sequelae. Among the intra-alveolar and peri-bronchiolar lymphocyte infiltrates, human lung-resident (CD103+) CD8+ and CD4+ T cells were sequestered in epithelial (CD326+) lung niches and secreted granzyme B and perforin, suggesting anti-viral cytotoxic activity. Infected mice also mounted human IgG antibody responses to SARS-CoV-2 viral proteins. Hence, HIS-DRAGA mice showed unique advantages as a surrogate in vivo human model for studying SARS-CoV-2 immunopathological mechanisms and testing the safety and efficacy of candidate vaccines and therapeutics.


Asunto(s)
COVID-19 , Antígeno HLA-DR4 , Animales , Linfocitos B , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Células Endoteliales , Antígeno HLA-A2/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , SARS-CoV-2
6.
Eur J Immunol ; 40(8): 2277-88, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20540111

RESUMEN

Polyclonality of self-reactive CD4(+) T cells is the hallmark of several autoimmune diseases like type 1 diabetes. We have previously reported that a soluble dimeric MHC II-peptide chimera prevents and reverses type 1 diabetes induced by a monoclonal diabetogenic T-cell population in double Tg mice [Casares, S. et al., Nat. Immunol. 2002. 3: 383-391]. Since most of the glutamic acid decarboxylase 65 (GAD65)-specific CD4(+) T cells in the NOD mouse are tolerogenic but unable to function in an autoimmune environment, we have activated a silent, monoclonal T-regulatory cell population (GAD65(217-230)-specific CD4(+) T cells) using a soluble I-A(αß) (g7)/GAD65(217-230)/Fcγ2a dimer, and measured the effect on the ongoing polyclonal diabetogenic T-cell process. Activated GAD65(217-230)-specific T cells and a fraction of the diabetogenic (B(9-23)-specific) T cells were polarized toward the IL-10-secreting T-regulatory type 1-like function in the pancreas of diabetic NOD mice. More importantly, this led to the reversal of hyperglycemia for more than 2 months post-therapy in 80% of mice in the context of stabilization of pancreatic insulitis and improved insulin secretion by the ß cells. These findings argue for the stabilization of a polyclonal self-reactive T-cell process by a single epitope-mediated bystander suppression. Dimeric MHC class II-peptide chimeras-like approach may provide rational grounds for the development of more efficient antigen-specific therapies in type 1 diabetes.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Interleucina-10/biosíntesis , Páncreas/patología , Linfocitos T Reguladores/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Células Clonales , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 1/terapia , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Hiperglucemia , Terapia de Inmunosupresión , Insulina/genética , Insulina/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
7.
J Immunol ; 182(1): 102-10, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109140

RESUMEN

Naturally occurring CD4(+)25(high)Foxp3(+) T regulatory (T-reg) cells are critical for maintaining tolerance to self and non-self Ags. The Foxp3 master-regulatory gene and CD28 costimulation are both required for thymic development and suppressogenic function of CD4(+)25(high)Foxp3(+) T-regs. Herein, we show that the sole CD28 stimulation of T-reg thymic precursors augments Foxp3 expression through the increase in Foxp3 mRNA life span by a mechanism involving p56(lck) and its binding motif on CD28 cytosolic tail, as well as the lipid rafts. We found that 1) the glycosphingolipids and cholesterol components of lipid rafts were highly expressed and unusually partitioned in T-reg thymic precursors as compared with the conventional T cell precursors, 2) the CD28 receptor density on cell membrane is proportional with the content of cholesterol in lipid rafts and with the level of Foxp3 mRNA expression in T-reg precursors, and 3) the CD28-mediated increase of Foxp3 mRNA life span was paralleled by an increased proliferative and suppressogenic capacity of terminally differentiated CD4(+)25(high)Foxp3(+) T-reg precursors. Thus, the functional integrity of CD28 receptor p56(lck) and plasma membrane lipid rafts are all prerequisites for up-regulation and long-term expression of Foxp3 mRNA transcripts in CD4(+)25(high)Foxp3(+) T-reg precursors.


Asunto(s)
Antígenos CD28/fisiología , Factores de Transcripción Forkhead/genética , Microdominios de Membrana/inmunología , Estabilidad del ARN/inmunología , Transducción de Señal/inmunología , Células Madre/inmunología , Linfocitos T Reguladores/inmunología , Familia-src Quinasas/fisiología , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/fisiología , Secuencias de Aminoácidos , Animales , Antígenos CD28/genética , Ciclo Celular/genética , Ciclo Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Citosol/enzimología , Citosol/inmunología , Factores de Transcripción Forkhead/metabolismo , Microdominios de Membrana/enzimología , Microdominios de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Unión Proteica/genética , Unión Proteica/inmunología , Estabilidad del ARN/genética , ARN Mensajero/biosíntesis , Transducción de Señal/genética , Células Madre/enzimología , Células Madre/metabolismo , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/metabolismo , Timo/citología , Timo/enzimología , Timo/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Familia-src Quinasas/deficiencia , Familia-src Quinasas/genética
8.
bioRxiv ; 2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-32839773

RESUMEN

We report the first Human Immune System (HIS)-humanized mouse model ("DRAGA": HLA-A2.HLA-DR4.Rag1KO.IL-2RγcKO.NOD) for COVID-19 research. This mouse is reconstituted with human cord blood-derived, HLA-matched hematopoietic stem cells. It engrafts human epi/endothelial cells expressing the human ACE2 receptor for SARS-CoV-2 and TMPRSS2 serine protease co-localized on lung epithelia. HIS-DRAGA mice sustained SARS-CoV-2 infection, showing deteriorated clinical condition, replicating virus in the lungs, and human-like lung immunopathology including T-cell infiltrates, microthrombi and pulmonary sequelae. Among T-cell infiltrates, lung-resident (CD103+) CD8+ T cells were sequestered in epithelial (CD326+) lung niches and secreted granzyme B and perforin, indicating cytotoxic potential. Infected mice also developed antibodies against the SARS-CoV-2 viral proteins. Hence, HIS-DRAGA mice showed unique advantages as a surrogate in vivo human model for studying SARS-CoV-2 immunopathology and for testing the safety and efficacy of candidate vaccines and therapeutics.

9.
Hum Vaccin Immunother ; 16(9): 2222-2237, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32129705

RESUMEN

We have engineered a Human Immune System (HIS)-reconstituted mouse strain (DRAGA mouse: HLA-A2. HLA-DR4. Rag1 KO. IL-2Rγc KO. NOD) in which the murine immune system has been replaced by a long-term, functional HIS via infusion of CD34+ hematopoietic stem cells (HSC) from cord blood. Herein, we report that the DRAGA mice can sustain inducible and transmissible H1N1 and H3N2 influenza A viral (IAV) infections. DRAGA female mice were significantly more resilient than the males to the H3N2/Aichi infection, but not to H3N2/Hong Kong, H3N2/Victoria, or H1N1/PR8 sub-lethal infections. Consistently associated with large pulmonary hemorrhagic areas, both human and murine Factor 8 mRNA transcripts were undetectable in the damaged lung tissues but not in livers of DRAGA mice advancing to severe H1N1/PR8 infection. Infected DRAGA mice mounted a neutralizing anti-viral antibody response and developed lung-resident CD103 T cells. These results indicate that the DRAGA mouse model for IAV infections can more closely approximate the human lung pathology and anti-viral immune responses compared to non-HIS mice. This mouse model may also allow further investigations into gender-based resilience to IAV infections, and may potentially be used to evaluate the efficacy of IAV vaccine regimens for humans.


Asunto(s)
Modelos Animales de Enfermedad , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Femenino , Antígeno HLA-A2/genética , Antígenos HLA-C , Antígeno HLA-DR4 , Proteínas de Homeodominio , Hong Kong , Humanos , Subtipo H3N2 del Virus de la Influenza A , Pulmón , Ratones , Ratones Endogámicos NOD
10.
Transplantation ; 85(12): 1717-25, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18580462

RESUMEN

BACKGROUND: Transplantation of pancreatic islets showed a tremendous progress over the years as a promising, new therapeutic strategy in patients with type 1 diabetes. However, additional immunosuppressive drug therapy is required to prevent rejection of engrafted islets. The current immunosuppressive therapies showed limited success in maintaining long-term islet survival as required to achieve insulin independence in type 1 diabetes, and they induce severe adverse effects. Herein, we analyzed the effects of a soluble peptide-major histocompatibility complex (MHC) class II chimera aimed at devising an antigen-specific therapy for suppression of anti-islet T cell responses and to improve the survival of pancreatic islets transplants. METHODS: Pancreatic islets from transgenic mice expressing the hemagglutinin antigen in the beta islets under the rat insulin promoter (RIP-HA) were grafted under the kidney capsule of diabetic, double transgenic mice expressing hemagglutinin in the pancreas and T cells specific for hemagglutinin (RIP-HA, TCR-HA). The recipient double transgenic mice were treated or not with the soluble peptide-MHC II chimera, and the progression of diabetes, graft survival, and T cell responses to the grafted islets were analyzed. RESULTS: The peptide-MHC II chimera protected syngeneic pancreatic islet transplants against the islet-reactive CD4 T cells, and prolonged the survival of transplanted islets. Protection of transplanted islets occurred by polarization of antigen-specific memory CD4 T cells toward a Th2 anti-inflammatory response. CONCLUSIONS: The peptide-MHC II chimera approach is an efficient and specific therapeutic approach to suppress anti-islet T cell responses and provides a long survival of pancreatic grafted islets.


Asunto(s)
Quimera/genética , Diabetes Mellitus Tipo 1/cirugía , Genes MHC Clase II/genética , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Péptidos/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Diabetes Mellitus Tipo 1/patología , Dimerización , Modelos Animales de Enfermedad , Hemaglutininas/genética , Hemaglutininas/metabolismo , Hiperglucemia/patología , Hiperglucemia/prevención & control , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Ratones , Ratones Transgénicos , Células Th2/inmunología , Células Th2/patología
11.
Mol Immunol ; 44(4): 530-40, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16597465

RESUMEN

The GM gangliosides and cholesterol components of plasma membrane lipid rafts play an important role in the recruitment and signaling of protein receptors in eukaryotic cells. Herein, we have analyzed at the single-cell level the partitioning and intracellular trafficking of GM gangliosides and cholesterol in quiescent (CD4+CD69-) and CD3-activated (CD4+CD69+) thymic and splenic T cells. First, regardless the gender and the quiescent or activated status of T cells, the GM and cholesterol content in cytosol and plasma membrane as well as the expression levels of GM synthase, Sphingomyelin phosphodiestarase 2 and HMG Co-A reductase genes involved in GM and cholesterol synthesis were constantly lower in CD4 thymocytes than in CD4 splenocytes. Second, we detected variations in the balance between GM and cholesterol in plasma membrane depending on aging, and found that deprivation of cellular cholesterol does not necessarily affect the GM content in both quiescent CD4 thymocytes and splenocytes. Third, CD3 stimulation up-regulated the GM and little if any the cholesterol content in both thymic and splenic CD4 T cells, suggesting a cross talk between the CD3 signaling and GM but not cholesterol biosynthesis pathway. Fourth, partitioning and trafficking of GM to the plasma membrane depended on the transport of ceramide precursors from endoplasmic reticulum to Golgi network, as well as on the synthesis, glycosylation and vesicular assembly in trans-Golgi, and less on the cytoskeleton architecture in both quiescent and activated CD4 thymic and splenic T cells. Together, these findings suggest that the differential partitioning and intracellular trafficking of GM and cholesterol in thymic and splenic CD4 T cells may account for the stage of functional maturation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Colesterol/metabolismo , Gangliósidos/metabolismo , Animales , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Transporte Biológico/genética , Transporte Biológico/inmunología , Complejo CD3 , Linfocitos T CD4-Positivos/inmunología , Balactosiltransferasa de Gangliósidos/biosíntesis , Balactosiltransferasa de Gangliósidos/genética , Regulación Enzimológica de la Expresión Génica/inmunología , Hidroximetilglutaril-CoA Reductasas/biosíntesis , Hidroximetilglutaril-CoA Reductasas/genética , Lectinas Tipo C , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Esfingomielina Fosfodiesterasa/biosíntesis , Esfingomielina Fosfodiesterasa/genética , Bazo/inmunología , Bazo/metabolismo , Timo/inmunología , Timo/metabolismo
12.
Hum Vaccin Immunother ; 14(2): 345-360, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29135340

RESUMEN

Pandemic outbreaks of influenza type A viruses have resulted in numerous fatalities around the globe. Since the conventional influenza vaccines (CIV) provide less than 20% protection for individuals with weak immune system, it has been considered that broadly cross-neutralizing antibodies may provide a better protection. Herein, we showed that a recently generated humanized mouse (DRAGA mouse; HLA-A2. HLA-DR4. Rag1KO. IL-2Rgc KO. NOD) that lacks the murine immune system and expresses a functional human immune system can be used to generate cross-reactive, human anti-influenza monoclonal antibodies (hu-mAb). DRAGA mouse was also found to be suitable for influenza virus infection, as it can clear a sub-lethal infection and sustain a lethal infection with PR8/A/34 influenza virus. The hu-mAbs were designed for targeting a human B-cell epitope (180WGIHHPPNSKEQ QNLY195) of hemagglutinin (HA) envelope protein of PR8/A/34 (H1N1) virus with high homology among seven influenza type A viruses. A single administration of HA180-195 specific hu-mAb in PR8-infected DRAGA mice significantly delayed the lethality by reducing the lung damage. The results demonstrated that DRAGA mouse is a suitable tool to (i) generate heterotype cross-reactive, anti-influenza human monoclonal antibodies, (ii) serve as a humanized mouse model for influenza infection, and (iii) assess the efficacy of anti-influenza antibody-based therapeutics for human use.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Infecciones por Orthomyxoviridae/terapia , Secuencia de Aminoácidos , Animales , Humanos , Virus de la Influenza A/clasificación , Virus de la Influenza A/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Pruebas de Neutralización , Conformación Proteica
13.
Malar Res Treat ; 2016: 6132734, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28074170

RESUMEN

Background. Plasmodium yoelii 17XNL is a nonlethal malaria strain in mice of different genetic backgrounds including the C57BL/6 mice (I-Ab/I-Enull) used in this study as a control strain. We have compared the trends of blood stage infection with the nonlethal murine strain of P. yoelii 17XNL malaria protozoan in immunocompetent Nonobese Diabetic (NOD) mice prone to type 1 diabetes (T1D) and C57BL/6 mice (control mice) that are not prone to T1D and self-cure the P. yoelii 17XNL infection. Prediabetic NOD mice could not mount a protective antibody response to the P. yoelii 17XNL-infected red blood cells (iRBCs), and they all succumbed shortly after infection. Our data suggest that the lack of anti-P. yoelii 17XNL-iRBCs protective antibodies in NOD mice is a result of parasite-induced, Foxp3+ T regulatory (Treg) cells able to suppress the parasite-specific antibody secretion. Conclusions. The NOD mouse model may help in identifying new mechanisms of B-cell evasion by malaria parasites. It may also serve as a more accurate tool for testing antimalaria therapeutics due to the lack of interference with a preexistent self-curing mechanism present in other mouse strains.

14.
Sci Rep ; 6: 28093, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27323875

RESUMEN

Humanized mice expressing Human Leukocyte Antigen (HLA) class I or II transgenes have been generated, but the role of class I vs class II on human T and B cell reconstitution and function has not been investigated in detail. Herein we show that NRG (NOD.RagKO.IL2RγcKO) mice expressing HLA-DR4 molecules (DRAG mice) and those co-expressing HLA-DR4 and HLA-A2 molecules (DRAGA mice) did not differ in their ability to develop human T and B cells, to reconstitute cytokine-secreting CD4 T and CD8 T cells, or to undergo immunoglobulin class switching. In contrast, NRG mice expressing only HLA-A2 molecules (A2 mice) reconstituted lower numbers of CD4 T cells but similar numbers of CD8 T cells. The T cells from A2 mice were deficient at secreting cytokines, and their B cells could not undergo immunoglobulin class switching. The inability of A2 mice to undergo immunoglobulin class switching is due to deficient CD4 helper T cell function. Upon immunization, the frequency and cytotoxicity of antigen-specific CD8 T cells in DRAGA mice was significantly higher than in A2 mice. The results indicated a multifactorial effect of the HLA-DR4 transgene on development and function of human CD4 T cells, antigen-specific human CD8 T cells, and immunoglobulin class switching.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Antígeno HLA-A2/genética , Antígeno HLA-DR4/genética , Animales , Linfocitos B/citología , Linfocitos T CD4-Positivos/citología , Citocinas/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina/genética , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Péptidos/síntesis química , Péptidos/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Proteínas Virales/síntesis química , Proteínas Virales/inmunología
15.
Mol Immunol ; 41(4): 399-409, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15163537

RESUMEN

The plasma membrane of T cells is made of a combination of glycosphingolipids and protein receptors organized in glycolipoprotein microdomains termed lipid rafts. The structural assembly of lipid rafts was investigated by various physical and biochemical assays. Depending on the differentiation status of T cells, the lipid rafts seclude various protein receptors involved in T cell signaling, cytoskeleton reorganization, membrane trafficking, and the entry of infectious organisms into the cells. This review article summarizes the most common methods, and their limits and advantages for analyzing the composition and assembly of lipid rafts with protein receptors into lipid rafts microdomains in plasma membrane of T cells. It also includes new methods such as ELISA/Polysorp and flow cytometry, and a combined sucrose gradient centrifugation-FPLC-Western blot strategy developed in our laboratory to study non-covalent interactions between the GM1 glycosphingolipid and protein receptors in plasma membrane of T cells.


Asunto(s)
Microdominios de Membrana/fisiología , Linfocitos T/ultraestructura , Animales , Western Blotting , Diferenciación Celular , Cromatografía en Capa Delgada , Citoesqueleto/ultraestructura , Citometría de Flujo , Glicoesfingolípidos/aislamiento & purificación , Glicoesfingolípidos/fisiología , Humanos , Ligandos , Sustancias Macromoleculares , Lípidos de la Membrana/aislamiento & purificación , Lípidos de la Membrana/fisiología , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/fisiología , Microscopía Fluorescente , Receptores de Superficie Celular/fisiología , Linfocitos T/inmunología
16.
Int Immunopharmacol ; 4(4): 521-6, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15099529

RESUMEN

Calicheamicin gamma1 (Cal gamma1) is a hydrophobic enediyne antibiotic known to cleave the DNA and lead to apoptosis in a variety of cells. Herein, we show that Cal gamma1 exhibits a 1000-times stronger suppressogenic effect on antigen-specific (diabetogenic), and naïve CD4 T cells than Doxorubicin (Dox), another strong apoptotic drug. The thymic precursors and mature T cells incubated with Cal gamma1 for only 30 min showed a drastic decrease or loss of cytokine production and proliferation following stimulation with the immunogenic peptide, or with CD3 and CD28 antibodies. The suppressogenicity of Cal gamma1 correlated with a rapid and non-selective degradation of RNA, whereas the DNA cleavage occurred at a later time point and at higher doses. Cal gamma1 may represent a potential therapeutic agent to eliminate self-reactive T cells in autoimmune diseases, providing that is delivered by antigen-specific T-cell ligands. Targeting of highly suppressogenic drugs such as Cal gamma1 to autoreactive T cells may reduce considerable the therapeutic dose and the drug-related side effects.


Asunto(s)
Aminoglicósidos/farmacología , Antibióticos Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , ADN/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , ADN/metabolismo , Doxorrubicina/farmacología , Enediinos , Ratones , Ratones Endogámicos BALB C
17.
Arch Immunol Ther Exp (Warsz) ; 52(4): 215-24, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15467486

RESUMEN

The plasma membrane of T cells is made up of a combination of phospholipids and proteins organized as glycolipoprotein microdomains termed lipid rafts. The structural assembly of lipid rafts was investigated by various physical and biochemical assays. Depending on the differentiation status of T cells, the lipid rafts seclude various protein receptors instrumental for the early T cell signaling, cytoskeleton reorganization, protein and membrane trafficking, and the entry of infectious organisms into the cells. This review article summarizes recent information on the assembly of lipid rafts in plasma membrane of T cells and their signaling output in mature and thymic precursors towards cell growth and differentiation, and possible modalities by which the function of lipid rafts can be altered by drugs and T cell ligands. The concept of using lipid rafts as a target for pharmaceutical compounds and biological T cell ligands to ultimately alter the T cell function is discussed.


Asunto(s)
Microdominios de Membrana/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Antígenos CD/metabolismo , Comunicación Celular/fisiología , Citoesqueleto/metabolismo , Humanos , Microdominios de Membrana/química , Proteínas de la Membrana/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/fisiología
18.
Rev Med Chir Soc Med Nat Iasi ; 118(2): 339-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25076697

RESUMEN

AIM: To determine the epidemiological and viroimmunological features and outcome of HIV/HBV-co infected patients cared in the lasi HIV/AIDS Regional Center. MATERIAL AND METHODS: This retrospective study included 252 patients diagnosed with HIV infection and associated hepatitis B virus (HBV) infection assessed at the Hospital of Infectious Diseases in the interval 2000-2013 and treated with antiretroviral drugs active against both HIV and HBV. RESULTS: The prevalence of HIV/HBV co infection was 19.9%. A slightly higher frequency of this co infection was found among males (53.2%); most patients belonged to age group 20-29 years (86.5%), mean age was 25.56 years. The predominant route of transmission was parenteral (58.5%), followed by heterosexual transmission (40.1%). The mean CD4 cell count was 246.20 cells/mm3, in over 41% of cases CD4 count ranging from 200 to 499 cells/mm3. The mean HIV plasma viral load was 142,906 copies/ml. ALT levels varied between 10-323 IU/l, average 49.90 IU/l, over 65% of subjects having pathological levels. In 21.8% of the cases, total cholesterol was very high, and in 16.8% of the patients the serum triglyceride levels were below the reference range (160 mg %). CONCLUSIONS: Our results suggest that HIV-positive patients, chronic hepatitis B infection has a high incidence, especially in younger age groups and is correlated with significant degrees of immunosuppression.


Asunto(s)
Coinfección , Infecciones por VIH/complicaciones , Infecciones por VIH/epidemiología , Hepatitis B/complicaciones , Hepatitis B/epidemiología , Adolescente , Adulto , Antirretrovirales/uso terapéutico , Niño , Femenino , Infecciones por VIH/diagnóstico , Infecciones por VIH/tratamiento farmacológico , Hepatitis B/diagnóstico , Hepatitis B/tratamiento farmacológico , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Estudios Retrospectivos , Factores de Riesgo , Rumanía/epidemiología , Distribución por Sexo
19.
Hum Vaccin Immunother ; 10(3): 693-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24335553

RESUMEN

Several human MHC class II (HLA) molecules are strongly associated with high incidence of autoimmune diseases including type 1 diabetes (T1D). The HLA-humanized mice may thus represent valuable tools to test HLA-based vaccines and therapeutics for human autoimmune diseases. Herein, we have tested the therapeutic potential of a soluble HLA-DR4-GAD65 271-280 (hu DEF-GAD65) chimera of human use in a newly-generated NOD/DR4/B7 double transgenic (dTg) mouse that develops spontaneously an accelerated T1D regardless the gender. The NOD/DR4/B7 dTg mice generated by a two-step crossing protocol express the HLA-DR*0401 molecules on 20% of antigen presenting cells, the human B7 molecules in pancreas, and HLA-DR4/GAD65-specific T-cells in the blood. Some 75% of pre-diabetic NOD/DR4/B7 dTg mice treated with hu DEF-GAD65 chimera remained euglycemic and showed a stabilized pancreatic insulitis 6 months after treatment. The 25% non responders developing hyperglycemia survived 3-4 months longer than their untreated littermates. T1D prevention by this reagent occurred by a Th2/TR-1 polarization in the pancreas. This study strongly suggests that the use of soluble pHLA reagents to suppress/stabilize the T1D progression and to extend the life expectancy in the absence of side effects is an efficient and safe therapeutic approach.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Glutamato Descarboxilasa/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Animales , Modelos Animales de Enfermedad , Glutamato Descarboxilasa/genética , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Longevidad , Ratones , Ratones SCID , Ratones Transgénicos , Páncreas/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Análisis de Supervivencia , Resultado del Tratamiento
20.
Front Syst Neurosci ; 8: 91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24999319

RESUMEN

Nanoscale structures have been at the core of research efforts dealing with integration of nanotechnology into novel electronic devices for the last decade. Because the size of nanomaterials is of the same order of magnitude as biomolecules, these materials are valuable tools for nanoscale manipulation in a broad range of neurobiological systems. For instance, the unique electrical and optical properties of nanowires, nanotubes, and nanocables with vertical orientation, assembled in nanoscale arrays, have been used in many device applications such as sensors that hold the potential to augment brain functions. However, the challenge in creating nanowires/nanotubes or nanocables array-based sensors lies in making individual electrical connections fitting both the features of the brain and of the nanostructures. This review discusses two of the most important applications of nanostructures in neuroscience. First, the current approaches to create nanowires and nanocable structures are reviewed to critically evaluate their potential for developing unique nanostructure based sensors to improve recording and device performance to reduce noise and the detrimental effect of the interface on the tissue. Second, the implementation of nanomaterials in neurobiological and medical applications will be considered from the brain augmentation perspective. Novel applications for diagnosis and treatment of brain diseases such as multiple sclerosis, meningitis, stroke, epilepsy, Alzheimer's disease, schizophrenia, and autism will be considered. Because the blood brain barrier (BBB) has a defensive mechanism in preventing nanomaterials arrival to the brain, various strategies to help them to pass through the BBB will be discussed. Finally, the implementation of nanomaterials in neurobiological applications is addressed from the brain repair/augmentation perspective. These nanostructures at the interface between nanotechnology and neuroscience will play a pivotal role not only in addressing the multitude of brain disorders but also to repair or augment brain functions.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA