Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Infect Dis ; 114: 58-61, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34757006

ABSTRACT

We describe a case of prolonged COVID-19 caused by the SARS-CoV-2 Gamma variant in a fully vaccinated healthcare worker, 387 days after an infection caused by lineage B.1.1.33. Infections were confirmed by whole-genome sequencing and corroborated by the detection of neutralizing antibodies in convalescent serum samples. Considering the permanent exposure of this healthcare worker to SARS-CoV-2, the waning immunity after the first infection, the low efficacy of the inactivated vaccine at preventing COVID-19, the immune escape of the Gamma variant (VOC), and the burden of post-COVID syndrome, this individual would have benefited from an additional dose of a heterologous vaccine.


Subject(s)
COVID-19 , SARS-CoV-2 , Brazil , COVID-19/complications , COVID-19/therapy , Humans , Immunization, Passive , Reinfection , Vaccines, Inactivated , COVID-19 Serotherapy , Post-Acute COVID-19 Syndrome
2.
J Infect Dis ; 224(6): 1060-1068, 2021 09 17.
Article in English | MEDLINE | ID: mdl-33528564

ABSTRACT

BACKGROUND: Zika virus (ZIKV) is associated with severe congenital abnormalities and laboratory diagnosis of antenatal infection is difficult. Here we evaluated ZIKV neutralizing antibody (nAb) kinetics in infants born to mothers with PCR-confirmed ZIKV infection during pregnancy. METHODS: Neonates (n = 98) had serum specimens tested repeatedly for ZIKV nAb over the first 2 years of life using virus neutralization test (VNT). ZIKV neonatal infection was confirmed by RT-PCR in blood or urine and/or presence of ZIKV IgM antibodies, and results were correlated with infant clinical features. RESULTS: Postnatal laboratory evidence of ZIKV vertical transmission was obtained for 60.2% of children, while 32.7% exhibited clinical abnormalities. Congenital abnormalities were found in 37.3% of children with confirmed ZIKV infection and 31.0% of children without confirmed infection (P = .734). All but 1 child displayed a physiologic decline in ZIKV nAb, reflecting maternal antibody decay, despite an early ZIKV-IgM response in one-third of infants. CONCLUSIONS: Infants with antenatal ZIKV exposure do not develop ZIKV nAb despite an early IgM response. Therefore, ZIKV VNT in children is not useful for diagnosis of congenital infection. In light of these findings, it remains to be determined if children infected in utero are potentially susceptible to reinfection.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Infectious Disease Transmission, Vertical , Pregnancy Complications, Infectious , Zika Virus Infection/diagnosis , Zika Virus/immunology , Biomarkers , Female , Humans , Immunoglobulin M , Infant , Infant, Newborn , Kinetics , Male , Polymerase Chain Reaction , Pregnancy , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/congenital
3.
Nat Commun ; 11(1): 3510, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665616

ABSTRACT

We report Zika virus (ZIKV) vertical transmission in 130 infants born to PCR+ mothers at the time of the Rio de Janeiro epidemic of 2015-2016. Serum and urine collected from birth through the first year of life were tested by quantitative reverse transcriptase polymerase chain reaction (PCR) and/or IgM Zika MAC-ELISA. Four hundred and seven specimens are evaluated; 161 sera tested by PCR and IgM assays, 85 urines by PCR. Sixty-five percent of children (N = 84) are positive in at least one assay. Of 94 children tested within 3 months of age, 70% are positive. Positivity declines to 33% after 3 months. Five children are PCR+ beyond 200 days of life. Concordance between IgM and PCR results is 52%, sensitivity 65%, specificity 40% (positive PCR results as gold standard). IgM and serum PCR are 61% concordant; serum and urine PCR 55%. Most children (65%) are clinically normal. Equal numbers of children with abnormal findings (29 of 45, 64%) and normal findings (55 of 85, 65%) have positive results, p = 0.98. Earlier maternal trimester of infection is associated with positive results (p = 0.04) but not clinical disease (p = 0.98). ZIKV vertical transmission is frequent but laboratory confirmed infection is not necessarily associated with infant abnormalities.


Subject(s)
Communicable Diseases/transmission , Communicable Diseases/virology , Zika Virus Infection/transmission , Zika Virus Infection/virology , Zika Virus/pathogenicity , Female , Humans , Immunoglobulin M/metabolism , Polymerase Chain Reaction , Pregnancy , Virus Diseases/virology
4.
Crit Rev Immunol ; 40(3): 237-246, 2020.
Article in English | MEDLINE | ID: mdl-33389887

ABSTRACT

The autoimmune disease multiple sclerosis (MS) is driven by T cells that are reactive to self-antigens of the brain and spinal cord. Many drugs have been developed to treat MS, but we believe that immune-specific targeting of pathogenic T cells may be a better approach for treatment. This type of therapy identifies specific components of the self-reactive T-cell repertoire that would undergo similar natural selection criteria as those found in driver genes in cancer genesis. In the context of autoimmunity, we propose that a focused subpopulation of T cells "drive" disease and could be found in higher frequency and become over-represented during disease induction and subsequent MS relapses. In addition, identification of other key signatures of driver T cells is important. One such marker could be interleukin (IL)-17- producing T cells. Here, we discuss the use of experimental autoimmune encephalomyelitis (EAE) animal models (that mimic many pathologic mechanisms involved in MS) to identify possible driver clones of this autoimmunity within the set of T cells expressing the IL-17 cytokine. EAE can be induced by myelin injection-associated proteins in adjuvants. The disease model in the Swiss/Jackson laboratory mouse strain represents the most common form of MS in humans: relapsing remitting MS. Finally, we discuss the concept of using IL-17 as a marker for pathogenic T cells, combined with identifying their T-cell receptor V repertoire, which could provide targeted approaches designed to neutralize driver T cells for MS immunotherapy.


Subject(s)
Clone Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunotherapy, Adoptive/methods , Multiple Sclerosis, Relapsing-Remitting/therapy , Th17 Cells/immunology , Animals , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , High-Throughput Nucleotide Sequencing , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Th17 Cells/metabolism
6.
Nat Med ; 25(8): 1213-1217, 2019 08.
Article in English | MEDLINE | ID: mdl-31285631

ABSTRACT

We report neurodevelopmental outcomes in 216 infants followed since the time of PCR-confirmed maternal Zika virus (ZIKV) infection in pregnancy during the Rio de Janeiro epidemic of 2015-2016 (refs. 1,2). Neurodevelopment was assessed by Bayley Scales of Infant and Toddler Development, third edition (Bayley-III; cognitive, language and motor domains) in 146 children and through neurodevelopment questionnaires/neurological examinations in 70 remaining children. Complete eye exams (n = 137) and hearing assessments (n = 114) were also performed. Below-average neurodevelopment and/or abnormal eye or hearing assessments were noted in 31.5% of children between 7 and 32 months of age. Among children assessed by Bayley-III, 12% scored below -2 s.d. (score <70; a score of 100 ± 2 s.d. is the range) in at least one domain; and 28% scored between -1 and -2 s.d. in any domain (scores <85-70). Language function was most affected, with 35% of 146 children below average. Improved neurodevelopmental outcomes were noted in female children, term babies, children with normal eye exams and maternal infection later in pregnancy (P = 0.01). We noted resolution of microcephaly with normal neurodevelopment in two of eight children, development of secondary microcephaly in two other children and autism spectrum disorder in three previously healthy children in the second year of life.


Subject(s)
Neurodevelopmental Disorders/etiology , Sensation Disorders/etiology , Zika Virus Infection/congenital , Zika Virus Infection/complications , Adult , Autism Spectrum Disorder/etiology , Child, Preschool , Female , Hearing , Humans , Infant , Infant, Newborn , Male , Microcephaly/etiology , Pregnancy , Pregnancy Complications, Infectious , Prospective Studies , Vision, Ocular
7.
Clin Infect Dis ; 65(6): 877-883, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28535184

ABSTRACT

BACKGROUND: Congenital Zika virus (ZIKV) syndrome is a newly identified condition resulting from infection during pregnancy. We analyzed outcome data from a mother-infant cohort in Rio de Janeiro in order to assess whether clinical severity of maternal ZIKV infection was associated with maternal virus load, prior dengue antibodies, or abnormal pregnancy/infant outcomes. METHODS: A clinical severity assessment tool was developed based on duration of fever, severity of rash, multisystem involvement, and duration of symptoms during ZIKV infection. ZIKV-RNA load was quantified by polymerase chain reaction (PCR) cycles in blood/ urine. Dengue immunoglobulin G (IgG) antibodies were measured at baseline. Adverse outcomes were defined as fetal loss or a live infant with grossly abnormal clinical or brain imaging findings. Regression models were used to study potential associations. RESULTS: 131 ZIKV-PCR positive pregnant women were scored for clinical disease severity, 6 (4.6%) had mild disease, 98 (74.8%) had moderate disease, and 27 (20.6%) severe manifestations of ZIKV infection. There were 58 (46.4%) abnormal outcomes with 9 fetal losses (7.2%) in 125 pregnancies. No associations were found between: disease severity and abnormal outcomes (P = .961; odds ratio [OR]: 1.00; 95% confidence interval [CI]: 0.796-1.270); disease severity and viral load (P = .994); viral load and adverse outcomes (P = .667; OR: 1.02; 95% CI: 0.922-1.135); or existence of prior dengue antibodies (88% subjects) with severity score, ZIKV-RNA load or adverse outcomes (P = .667; OR: 0.78; 95% CI: 0.255-2.397). CONCLUSIONS: Congenital ZIKV syndrome does not appear to be associated with maternal disease severity, ZIKV-RNA load at time of infection or existence of prior dengue antibodies.


Subject(s)
Fetal Death , Nervous System Diseases/epidemiology , Nervous System Malformations/epidemiology , Pregnancy Complications, Infectious/blood , Zika Virus Infection/blood , Zika Virus Infection/complications , Adolescent , Adult , Antibodies, Viral/blood , Brain/abnormalities , Brain/diagnostic imaging , Brazil/epidemiology , Dengue Virus/immunology , Female , Humans , Live Birth/epidemiology , Middle Aged , Nervous System Diseases/congenital , Nervous System Diseases/diagnosis , Nervous System Diseases/physiopathology , Nervous System Malformations/diagnosis , Neuroimaging , Neurologic Examination , Pregnancy , Pregnancy Complications, Infectious/virology , Prospective Studies , RNA, Viral/blood , Severity of Illness Index , Viral Load , Young Adult , Zika Virus/genetics
8.
Curr Opin Pediatr ; 29(1): 107-113, 2017 02.
Article in English | MEDLINE | ID: mdl-27870689

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to present what is known about the Zika virus (ZIKV) at the time of writing this review. The viral structure and its phylogeny, as well as the limitations of current available techniques used for diagnosis, are discussed. RECENT FINDINGS: Crystallography and cryo-electron microscopy of the whole ZIKV, or a few of its proteins, are confirming its overall antigenic relatedness to other flaviviruses. Sequencing has revealed its dynamic genetic variation and has placed the Western cluster of Zika isolates within the Asian phylogenic tree. Genetic codon mutations, although highly prevalent, do not usually translate into modifications at amino acid or proteomic levels, revealing conserved enzymatic functions that could potentially be addressed therapeutically. Clinical characterization of ZIKV infection is complicated because of symptoms similar to dengue and chikungunya. Diagnosis requires specialized laboratories with costly reagents and highly trained personnel. Although commercial labs are now offering ZIKV diagnostic tests, most of them are not fully tested in comparison with standard molecular techniques standardized at CDC and local health departments. We are still in desperate need of simpler diagnostic tests that better discriminate ZIKV from coendemic arboviruses. SUMMARY: The area of better Zika diagnostic assays is a rapidly developing field with the public attention directed to this epidemic. Academic interest in this topic is driving fast disclosure of information in peer-reviewed journals and grey papers via web-based forums. We expect in the near future that new promising strategies for improved Zika diagnostics will translate into preventive and therapeutic tools.


Subject(s)
Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , DNA, Viral/analysis , Humans , Phylogeny , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/virology
9.
J Transl Med ; 9: 101, 2011 Jul 02.
Article in English | MEDLINE | ID: mdl-21722394

ABSTRACT

BACKGROUND: Non Obese Diabetic mice lacking B cells (NOD.Igµ(null) mice) do not develop diabetes despite their susceptible background. Upon reconstitution of B cells using a chimera approach, animals start developing diabetes at 20 weeks of age. METHODS: We have used the spectratyping technique to follow the T cell receptor (TCR) V beta repertoire of NOD.Igµ(null) mice following B cell reconstitution. This technique provides an unbiased approach to understand the kinetics of TCR expansion. We have also analyzed the TCR repertoire of reconstituted animals receiving cyclophosphamide treatment and following tissue transplants to identify common aggressive clonotypes. RESULTS: We found that B cell reconstitution of NOD.Igµ(null) mice induces a polyclonal TCR repertoire in the pancreas 10 weeks later, gradually diversifying to encompass most BV families. Interestingly, these clonotypic BV expansions are mainly confined to the pancreas and are absent from pancreatic lymph nodes or spleens. Cyclophosphamide-induced diabetes at 10 weeks post-B cell reconstitution reorganized the predominant TCR repertoires by removing potential regulatory clonotypes (BV1, BV8 and BV11) and increasing the frequency of others (BV4, BV5S2, BV9, BV16-20). These same clonotypes are more frequently present in neonatal pancreatic transplants under the kidney capsule of B-cell reconstituted diabetic NOD.Igµ(null) mice, suggesting their higher invasiveness. Phenotypic analysis of the pancreas-infiltrating lymphocytes during diabetes onset in B cell reconstituted animals show a predominance of CD19+ B cells with a B:T lymphocyte ratio of 4:1. In contrast, in other lymphoid organs (pancreatic lymph nodes and spleens) analyzed by FACS, the B:T ratio was 1:1. Lymphocytes infiltrating the pancreas secrete large amounts of IL-6 and are of Th1 phenotype after CD3-CD28 stimulation in vitro. CONCLUSIONS: Diabetes in NOD.Igµ(null) mice appears to be caused by a polyclonal repertoire of T cell accumulation in pancreas without much lymphoid organ involvement and is dependent on the help by B cells.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Immunoglobulin mu-Chains/immunology , Immunophenotyping/methods , Islets of Langerhans/immunology , T-Lymphocytes/immunology , Animals , Animals, Newborn , B-Lymphocytes/cytology , Cell Proliferation , Clone Cells , Cyclophosphamide , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Graft Rejection/complications , Graft Rejection/immunology , Graft Rejection/pathology , Immunologic Memory/immunology , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Mice , Mice, Inbred NOD , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/pathology , T-Lymphocytes/cytology
10.
J Transl Med ; 8: 98, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20946663

ABSTRACT

BACKGROUND: Tumor immune responses are first generated and metastases often begin in tumor sentinel lymph nodes (TSLN). Therefore, it is important to promote tumor immunity within this microenvironment. Mifepristone (RU486) treatment can interfere with cortisol signaling that can lead to suppression of tumor immunity. Here, we assessed whether treatment with RU486 in conjunction with an intratumor injection of Ad5IL-12 vector (a recombinant adenovirus expressing IL-12) could impact the TSLN microenvironment and prostate cancer progression. METHODS: The human PC3, LNCaP or murine TRAMP-C1 prostate cancer cell lines were used to generate subcutaneous tumors in NOD.scid and C57BL/6 mice, respectively. Adjuvant effects of RU486 were looked for in combination therapy with intratumor injections (IT) of Ad5IL-12 vector in comparison to PBS, DL70-3 vector, DL70-3 + RU486, RU486 and Ad5IL-12 vector treatment controls. Changes in tumor growth, cell cytotoxic activity and populations of CD4+/FoxP3+ T regulatory cells (Treg) in the TSLN were evaluated. RESULTS: Treatment of human PC3 prostate xenograft or TRAMP-C1 tumors with combination Ad5IL-12 vector and RU486 produced significantly better therapeutic efficacy in comparison to controls. In addition, we found that combination therapy increased the capacity of TSLN lymphocytes to produce Granzyme B in response to tumor cell targets. Finally, combination therapy tended towards decreases of CD4+/FoxP3+ T regulatory cell populations to be found in the TSLN. CONCLUSION: Inclusion of RU486 may serve as a useful adjuvant when combined with proinflammatory tumor killing agents by enhancement of the immune response and alteration of the TSLN microenvironment.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Interleukin-12/administration & dosage , Lymphatic Metastasis , Mifepristone/therapeutic use , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-12/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
11.
Mol Immunol ; 46(3): 366-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19042022

ABSTRACT

Following Leishmania major infection, the early LACK (Leishmania homolog of receptors for activated C kinase)-induced IL-4 response appears to determine disease susceptibility in BALB/c mice. Therefore, we sought to manipulate the pathogenic T cell responses to the immunodominant epitope with the use of altered peptide ligands (APLs). Conservative and non-conservative substitutions for each amino acid of the LACK 161-175 peptide determinant were tested for their stimulatory capacity in four different LACK-reactive T cell systems. From these results, we propose a likely LACK 163-171/I-A(d) core peptide register and show that APLs with changes at putative T cell receptor (TCR) contacts provide the greatest potential for immune deviation. In particular, the TCR-contact H164V APL expanded Th1 cells upon in vitro recall of naïve splenocytes from LACK-specific BV4 T cell receptor transgenic mice and stimulated IFN-gamma secretion from a Th2-committed LACK-reactive T cell line. We also observed that non-conservative substitutions flanking the core determinant had strong agonistic effects for proliferation and Th1/Th2 modulation. However, upon immunization, the H164V APL considerably downregulated proliferation and cytokine responses to the wild type LACK 161-175 peptide, while immunization with the weak agonist, MHC contact APL S171K, increased the IFN-gamma/IL-4 ratio to the wild type peptide. In these instances, a hyporesponsive T cell response to the wild-type peptide was achieved by immunizing with an APL possessing non-conservative substitutions at TCR contact sites, while immune deviation was accomplished using a weak-agonist APL that retained the core determinant. Thus, certain LACK-APLs are able to induce T cell responses with a protective phenotype in an infectious disease such as leishmaniasis.


Subject(s)
Antigens, Protozoan/immunology , Immunodominant Epitopes/immunology , Leishmania major/immunology , Peptides/immunology , Protozoan Proteins/immunology , Th2 Cells/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigens, Protozoan/chemistry , Cell Line , Cell Proliferation , Cross-Priming/immunology , Cytokines , Immunization , Interferon-gamma/immunology , Interleukin-4/immunology , Ligands , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Peptides/chemistry , Protozoan Proteins/chemistry , Protozoan Vaccines/immunology , Receptors, Antigen, T-Cell/immunology , Th2 Cells/cytology
12.
Vaccine ; 23(2): 247-57, 2004 Nov 25.
Article in English | MEDLINE | ID: mdl-15531044

ABSTRACT

Immunization with soluble leishmanial antigen (SLA) in IFA plus Ad5IL-12 vector induced protection confined to the immunized footpad in BALB/c mice. However, animals that controlled a primary infection with a Leishmania major challenge in the same immunized footpad, became resistant to subsequent contralateral rechallenges due to expansion of IFN-gamma secreting cells. This systemic immunity could be disrupted either by macrophage depletion during immunization or by lymphadenectomy after challenge. We show that this procedure does not interfere with tissue-compartmentalized protection, since lymphadenectomized and splenectomized animals were resistant to rechallenges performed in the immunized footpads. Our results indicate that SLA-Ad5IL-12 vector priming requires macrophages to generate systemic protection. Furthermore, a previously undescribed lymphoid organ-independent, protective immune response is contained within the tissue microenvironment of the immunized/challenged footpad. These results have important implications for vaccine design against leishmanial and mycobacterial infections and diseases caused by intracellular pathogens.


Subject(s)
Interleukin-12/therapeutic use , Leishmania major/immunology , Leishmaniasis, Cutaneous/prevention & control , Lymph Nodes/immunology , Macrophages/immunology , Adenoviridae/genetics , Animals , Antigens, Protozoan/immunology , Cell Line , Genetic Vectors/administration & dosage , Interleukin-12/genetics , Leishmania major/genetics , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Macrophages/drug effects , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/immunology
13.
J Immunol ; 172(6): 3447-53, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004144

ABSTRACT

To study the T cell responses induced by native and modified Ag, we have followed in vivo TCR selection and cytokine profile of T cells, as well as the isotype of induced Abs, in response to the model Ag hen egg-white lysozyme (HEL) and its reduced and carboxymethylated form (RCM-HEL). RCM-HEL induces in vivo a T cell response focused on the same immunodominant determinant characterizing the response to native HEL, but further skewed to the Th1 pathway. No difference between HEL and RCM-HEL could be observed in the efficiency of processing, nor in the type of APCs involved. In vivo experiments show that coimmunization with HEL and RCM-HEL generates distinct Th2 or Th1 responses in naive mice, but the two forms of Ag expand the same HEL-specific public clonotype, characterized by the Vbeta8.2-Jbeta1.5 rearrangement, indicating that the populations of naive T cells activated by the two Ag forms overlap. T cells primed by RCM-HEL are restimulated by soluble HEL in vivo, but divert the phenotype of the HEL-specific response to Th1, implying that priming of naive T cells by a structurally modified Ag can induce Th1-type memory/effector T cells more efficiently than native Ag.


Subject(s)
Antigens/immunology , Interphase/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens/administration & dosage , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Clone Cells , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/immunology , Female , Immunization , Immunodominant Epitopes/biosynthesis , Immunoglobulin Isotypes/biosynthesis , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred CBA , Muramidase/administration & dosage , Muramidase/immunology , Muramidase/metabolism , Oxidation-Reduction , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/cytology , Th1 Cells/immunology , Th1 Cells/metabolism
14.
J Autoimmun ; 22(1): 13-20, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14709409

ABSTRACT

Nasal installation or oral feeding of antigens can alter the subsequent immune response in animals and humans. Most mucosal treatments with antigens tend to down-regulate disease, inducing full tolerance or immune deviation; however, priming has also been reported. We evaluated the course of experimental autoimmune encephalomyelitis (EAE) in (SJL x B10.PL)F1 mice after nasal instillation of myelin basic protein. There was a tendency towards exacerbation of subsequent disease in animals if they were nasally exposed to gpMBP during the neonatal period (first week of life), compared to exposure during adulthood. Later, at 11 months of age, this tendency to exacerbate disappeared. Our results suggest that mucosal exposure during early life may regularly modulate the anti-self immune response upwards in individuals genetically predisposed to autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity, Mucosal , Myelin Basic Protein/immunology , Administration, Intranasal , Age Factors , Animals , Animals, Newborn , Down-Regulation , Guinea Pigs , Humans , Immunization, Secondary , Mice , Myelin Basic Protein/administration & dosage , Secondary Prevention , Th1 Cells/immunology , Th2 Cells/immunology
15.
Isr Med Assoc J ; 4(11 Suppl): 902-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12455176

ABSTRACT

BACKGROUND: Nasal instillation is an effective method for inducing antigen-specific immune tolerance. However, it is not clear how a tolerization scheme established in one mouse strain will perform when used in a mouse of a different haplotype. OBJECTIVES: To compare the antigen-specific recall responses in four mouse strains--BALB/c, C57BL/6, NOD, and B10.PL--that were pretreated nasally with 50 micrograms of hen egg-white lysozyme prior to parenteral immunization with homologous antigen. METHODS: Mice were nasally treated with a prototype antigen, HEL, and then immunized with the same antigen emulsified in complete Freund's adjuvant. Spleens and lymph nodes were assayed for T cell proliferation measured by tritiated thymidine incorporation. Cytokine production was measured using ELISPOT assay. Serum antibody response to HEL was measured using an enzyme-linked immunosorbent assay. RESULTS: Proliferative recall responses to HEL in B10.PL, C57BL/6, and BALB/c were greatly reduced compared to control mice, but non-obese diabetic mice were resistant to the tolerization regime. Despite their susceptibility to nasally induced suppression, the mechanisms responsible for tolerance induction differed in BALB/c and C57BL/6 mice. CONCLUSIONS: Our findings demonstrate that while mucosal contacts with specific antigen consistently affect the outcome of subsequent exposure to the same antigen, the observed response will vary non-predictably, depending on the genetic background of the animal.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Disease Models, Animal , Immune Tolerance/immunology , Immunization/methods , Mice, Inbred BALB C/immunology , Mice, Inbred C57BL/immunology , Mice, Inbred NOD/immunology , Muramidase/immunology , Muramidase/therapeutic use , Animals , Autoimmune Diseases/chemically induced , Drug Evaluation, Preclinical , Epitopes/genetics , Epitopes/immunology , Haplotypes/genetics , Haplotypes/immunology , Immune Tolerance/genetics , Immunity, Mucosal/immunology , Instillation, Drug , Lymphocyte Activation , Mice , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL/genetics , Mice, Inbred NOD/genetics , Nasal Mucosa , T-Lymphocytes/immunology
16.
Folha méd ; 98(6): 389-90, 1989. tab
Article in Portuguese | LILACS | ID: lil-172241

ABSTRACT

A ubiquinona (coenzima Q(10)) é um potente agente antioxidante, que tem sido usado como droga para a isquemia do miocárdio, já que reduz a injúria causada pela hipóxia. Como os radicais livres aumentam a agregaçao e a adesividade plaquetárias, estudamos a açao desta substância, por via sublingual, sobre a adesividade das plaquetas, usando o método in vivo de Borchgrevink. Verificou-se uma poderosa açao inibidora, o que sugere uma possível açao terapêutica da ubiquinona como agente de antiadesao das plaquetas.


Subject(s)
Humans , Adult , Middle Aged , Platelet Adhesiveness , Ubiquinone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...