Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Front Immunol ; 14: 1085922, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36865536

RESUMO

IgA nephropathy (IgAN) is an autoimmune disease in which poorly galactosylated IgA1 is the antigen recognized by naturally occurring anti-glycan antibodies, leading to formation of nephritogenic circulating immune complexes. Incidence of IgAN displays geographical and racial disparity: common in Europe, North America, Australia, and east Asia, uncommon in African Americans, many Asian and South American countries, Australian Aborigines, and rare in central Africa. In analyses of sera and cells from White IgAN patients, healthy controls, and African Americans, IgAN patients exhibited substantial enrichment for IgA-expressing B cells infected with Epstein-Barr virus (EBV), leading to enhanced production of poorly galactosylated IgA1. Disparities in incidence of IgAN may reflect a previously disregarded difference in the maturation of the IgA system as related to the timing of EBV infection. Compared with populations with higher incidences of IgAN, African Americans, African Blacks, and Australian Aborigines are more frequently infected with EBV during the first 1-2 years of life at the time of naturally occurring IgA deficiency when IgA cells are less numerous than in late childhood or adolescence. Therefore, in very young children EBV enters "non-IgA" cells. Ensuing immune responses prevent infection of IgA B cells during later exposure to EBV at older ages. Our data implicate EBV-infected cells as the source of poorly galactosylated IgA1 in circulating immune complexes and glomerular deposits in patients with IgAN. Thus, temporal differences in EBV primo-infection as related to naturally delayed maturation of the IgA system may contribute to geographic and racial variations in incidence of IgAN.


Assuntos
Infecções por Vírus Epstein-Barr , Glomerulonefrite por IGA , Adolescente , Criança , Pré-Escolar , Humanos , Complexo Antígeno-Anticorpo , Austrália , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/etnologia , Glomerulonefrite por IGA/epidemiologia , Glomerulonefrite por IGA/etnologia , Herpesvirus Humano 4 , Imunoglobulina A , População Negra , Lactente
2.
Front Immunol ; 13: 957107, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36059541

RESUMO

SARS-CoV-2 is primarily an airborne infection of the upper respiratory tract, which on reaching the lungs causes the severe acute respiratory disease, COVID-19. Its first contact with the immune system, likely through the nasal passages and Waldeyer's ring of tonsils and adenoids, induces mucosal immune responses revealed by the production of secretory IgA (SIgA) antibodies in saliva, nasal fluid, tears, and other secretions within 4 days of infection. Evidence is accumulating that these responses might limit the virus to the upper respiratory tract resulting in asymptomatic infection or only mild disease. The injectable systemic vaccines that have been successfully developed to prevent serious disease and its consequences do not induce antibodies in mucosal secretions of naïve subjects, but they may recall SIgA antibody responses in secretions of previously infected subjects, thereby helping to explain enhanced resistance to repeated (breakthrough) infection. While many intranasally administered COVID vaccines have been found to induce potentially protective immune responses in experimental animals such as mice, few have demonstrated similar success in humans. Intranasal vaccines should have advantage over injectable vaccines in inducing SIgA antibodies in upper respiratory and oral secretions that would not only prevent initial acquisition of the virus, but also suppress community spread via aerosols and droplets generated from these secretions.


Assuntos
COVID-19 , Imunidade nas Mucosas , Administração Intranasal , Animais , Humanos , Imunoglobulina A Secretora , Camundongos , SARS-CoV-2
3.
J Am Soc Nephrol ; 33(5): 908-917, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35115327

RESUMO

BACKGROUND: IgA nephropathy (IgAN) primary glomerulonephritis is characterized by the deposition of circulating immune complexes composed of polymeric IgA1 molecules with altered O-glycans (Gd-IgA1) and anti-glycan antibodies in the kidney mesangium. The mesangial IgA deposits and serum IgA1 contain predominantly λ light (L) chains, but the nature and origin of such IgA remains enigmatic. METHODS: We analyzed λ L chain expression in peripheral blood B cells of 30 IgAN patients, 30 healthy controls (HCs), and 18 membranous nephropathy patients selected as disease controls (non-IgAN). RESULTS: In comparison to HCs and non-IgAN patients, peripheral blood surface/membrane bound (mb)-Gd-IgA1+ cells from IgAN patients express predominantly λ L chains. In contrast, total mb-IgA+, mb-IgG+, and mb-IgM+ cells were preferentially positive for kappa (κ) L chains, in all analyzed groups. Although minor in comparison to κ L chains, λ L chain subsets of mb-IgG+, mb-IgM+, and mb-IgA+ cells were significantly enriched in IgAN patients in comparison to non-IgAN patients and/or HCs. In contrast to HCs, the peripheral blood of IgAN patients was enriched with λ+ mb-Gd-IgA1+, CCR10+, and CCR9+ cells, which preferentially home to the upper respiratory and digestive tracts. Furthermore, we observed that mb-Gd-IgA1+ cell populations comprise more CD138+ cells and plasmablasts (CD38+) in comparison to total mb-IgA+ cells. CONCLUSIONS: Peripheral blood of IgAN patients is enriched with migratory λ+ mb-Gd-IgA1+ B cells, with the potential to home to mucosal sites where Gd-IgA1 could be produced during local respiratory or digestive tract infections.


Assuntos
Glomerulonefrite por IGA , Feminino , Galactose , Humanos , Imunoglobulina A/metabolismo , Imunoglobulina G , Imunoglobulina M , Masculino
4.
Hum Vaccin Immunother ; 18(2): 1899549, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33939589

RESUMO

Extensive experiments performed mostly in a variety of animal models convincingly demonstrated the protective effect of polyclonal or monoclonal antibodies administered by the mucosal route. Because of the independence of the mucosal and systemic compartments of the immune system, antibodies from the circulation are not effectively transported in sufficient quantities into external secretions. Nevertheless, local application of antibodies of the desired specificity to mucosal membranes of the respiratory, gastrointestinal, and female genital tracts protected experimental animals from the subsequent challenge by corresponding viral or bacterial pathogens. Thus, generation of monoclonal antibodies of desired specificity and the selection of delivery systems to extend their otherwise short survival on some mucosal surfaces are essential aims of their usability in humans for the effective prevention of mucosally acquired infectious diseases.


Assuntos
Imunidade nas Mucosas , Mucosa , Animais , Anticorpos Monoclonais , Feminino , Trato Gastrointestinal , Sistema Imunitário
5.
Mucosal Immunol ; 14(2): 511-522, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32973324

RESUMO

Mucosal surfaces are colonized by highly diverse commensal microbiota. Coating with secretory IgA (SIgA) promotes the survival of commensal bacteria while it inhibits the invasion by pathogens. Bacterial coating could be mediated by antigen-specific SIgA recognition, polyreactivity, and/or by the SIgA-associated glycans. In contrast to many in vitro studies, only a few reported the effect of SIgA glycans in vivo. Here, we used a germ-free antibody-free newborn piglets model to compare the protective effect of SIgA, SIgA with enzymatically removed N-glycans, Fab, and Fc containing the secretory component (Fc-SC) during oral necrotoxigenic E. coli O55 challenge. SIgA, Fab, and Fc-SC were protective, whereas removal of N-glycans from SIgA reduced SIgA-mediated protection as demonstrated by piglets' intestinal histology, clinical status, and survival. In vitro analyses indicated that deglycosylation of SIgA did not reduce agglutination of E. coli O55. These findings highlight the role of SIgA-associated N-glycans in protection. Further structural studies of SIgA-associated glycans would lead to the identification of those involved in the species-specific inhibition of attachment to corresponding epithelial cells.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , Imunoglobulina A Secretora/metabolismo , Fragmentos Fab das Imunoglobulinas/metabolismo , Polissacarídeos/metabolismo , Anticorpos de Cadeia Única/metabolismo , Aglutinação , Animais , Animais Recém-Nascidos , Resistência à Doença , Feminino , Vida Livre de Germes , Glicosilação , Gravidez , Suínos
6.
Front Immunol ; 11: 611337, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329607

RESUMO

The mucosal immune system is the largest component of the entire immune system, having evolved to provide protection at the main sites of infectious threat: the mucosae. As SARS-CoV-2 initially infects the upper respiratory tract, its first interactions with the immune system must occur predominantly at the respiratory mucosal surfaces, during both inductive and effector phases of the response. However, almost all studies of the immune response in COVID-19 have focused exclusively on serum antibodies and systemic cell-mediated immunity including innate responses. This article proposes that there is a significant role for mucosal immunity and for secretory as well as circulating IgA antibodies in COVID-19, and that it is important to elucidate this in order to comprehend especially the asymptomatic and mild states of the infection, which appear to account for the majority of cases. Moreover, it is possible that mucosal immunity can be exploited for beneficial diagnostic, therapeutic, or prophylactic purposes.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19 , Imunidade nas Mucosas , Imunoglobulina A/imunologia , SARS-CoV-2/imunologia , COVID-19/diagnóstico , COVID-19/imunologia , COVID-19/patologia , Humanos
7.
Front Immunol ; 11: 267, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32184780

RESUMO

IgA nephropathy (IgAN) is the dominant type of primary glomerulonephritis worldwide. However, IgAN rarely affects African Blacks and is uncommon in African Americans. Polymeric IgA1 with galactose-deficient hinge-region glycans is recognized as auto-antigen by glycan-specific antibodies, leading to formation of circulating immune complexes with nephritogenic consequences. Because human B cells infected in vitro with Epstein-Barr virus (EBV) secrete galactose-deficient IgA1, we examined peripheral blood B cells from adult IgAN patients, and relevant controls, for the presence of EBV and their phenotypic markers. We found that IgAN patients had more lymphoblasts/plasmablasts that were surface-positive for IgA, infected with EBV, and displayed increased expression of homing receptors for targeting the upper respiratory tract. Upon polyclonal stimulation, these cells produced more galactose-deficient IgA1 than did cells from healthy controls. Unexpectedly, in healthy African Americans, EBV was detected preferentially in surface IgM- and IgD-positive cells. Importantly, most African Blacks and African Americans acquire EBV within 2 years of birth. At that time, the IgA system is naturally deficient, manifested as low serum IgA levels and few IgA-producing cells. Consequently, EBV infects cells secreting immunoglobulins other than IgA. Our novel data implicate Epstein-Barr virus infected IgA+ cells as the source of galactose-deficient IgA1 and basis for expression of relevant homing receptors. Moreover, the temporal sequence of racial-specific differences in Epstein-Barr virus infection as related to the naturally delayed maturation of the IgA system explains the racial disparity in the prevalence of IgAN.


Assuntos
Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/epidemiologia , Glomerulonefrite por IGA/virologia , Herpesvirus Humano 4/fisiologia , Grupos Raciais , República Tcheca/epidemiologia , Galactose , Glomerulonefrite por IGA/epidemiologia , Humanos , Imunoglobulina A/metabolismo , Lactente , Prevalência
8.
Artigo em Inglês | MEDLINE | ID: mdl-32176881

RESUMO

The human mammary gland is an integral effector component of the common mucosal immune system. However, from physiological and immunological aspects, it displays several unique features not shared by other mucosal sites. The development, maturation, and activity of the mammary gland exhibits a strong hormonal dependence. Furthermore, in comparison to the intestinal and respiratory tracts, the mammary gland is not colonized by high numbers of bacteria of enormous diversity and does not contain mucosal inductive sites analogous to the intestinal Peyer's patches. Consequently, when exposed to antigens, local or generalized immune responses are low or not present. Comparative evaluations of various immunization routes effective in the induction of antibodies in human milk are limited. Systemic immunization induces IgG antibodies in plasma, but due to the low levels of total IgG in human milk, their protective effect remains unknown. Oral or intranasal immunization or infection induces secretory IgA in milk, as demonstrated in several studies. Other routes of mucosal immunization, such as sublingual or rectal exposure effective in the induction of antibodies in various external secretions, have not been explored in the mammary gland. Because secretory IgA in milk displays protective functions, alternative immunization routes and antigen delivery systems should be explored.


Assuntos
Glândulas Mamárias Humanas , Leite Humano , Mucosa , Humanos , Sistema Imunitário , Imunoglobulina A Secretora , Leite Humano/imunologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-31723302

RESUMO

AIMS: Epstein-Barr virus (EBV) targets predominantly B cells and these cells could acquire new phenotype characteristics. Here we analyzed whether EBV-infected and -uninfected B cells from healthy subjects differ in proportion of dominant phenotypes, maturation stage, and homing receptors expression. METHODS: EBV-infected and -uninfected cells were identified by flow cytometry using fluorophore-labeled EBV RNA-specific DNA probes combined with fluorophore-labeled antibody to surface lineage markers, integrins, chemokine receptors, and immunoglobulin isotypes, including intracellular ones. RESULTS: Our results show that the trafficking characteristics of EBERpos B cells are distinct from EBERneg B cells with most dominant differences detected for α4ß1 and α4ß7 and CCR5 and CCR7. EBV-positive cells are predominantly memory IgM+ B cells or plasmablasts/plasma cells (PB/PC) positive for IgA or less for IgM. In comparison to uninfected B cells, less EBV-positive B cells express α4ß7 and almost no cells express α4ß1. EBV-positive B cells contained significantly higher proportion of CCR5+ and CCR7+ cells in comparison to EBV-negative cells. In vitro exposure of blood mononuclear cells to pro-inflammatory cytokine IL-6 reduces population of EBV-positive B cell. CONCLUSION: Although EBV-infected B cells represent only a minor subpopulation, their atypical functions could contribute in predisposed person to development abnormities such as some autoimmune diseases or tumors. Using multi-parameter flow cytometry we characterized differences in migration of EBV-positive and -negative B cells of various maturation stage and isotype of produced antibodies particularly different targeting to mucosal tissues of gastrointestinal and respiratory tracts.


Assuntos
Linfócitos B/imunologia , Sangue/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/fisiopatologia , Proteínas de Transporte Vesicular/imunologia , Proteínas de Transporte Vesicular/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Citometria de Fluxo , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade
10.
Curr Immunol Rev ; 15(1): 2-3, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-33223980
11.
Curr Immunol Rev ; 15(1): 41-48, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-33312087

RESUMO

The mucosal immune systems of the genital and intestinal tracts as the most frequent sites of HIV-1 entry, display remarkable immunological differences from the systemic immune compartment which must be considered in the evaluation of humoral and cellular immune responses to HIV-1. Marked differences in the fluids from the genital and intestinal tracts and in plasma with respect to the Ig isotypes, their levels, molecular forms and distinct effector functions must be taken into consideration in the evaluation and interpretation of humoral immune responses. Because of the low levels and highly pronounced variation in Ig content, HIV-1-specific antibody concentrations should be always related to the levels of total Ig of a given isotype. This practice will avoid inevitable differences due to the small volumes of collected fluids and sample dilution during the collection and processing of samples from external secretions. Furthermore, appropriate controls and immunochemical assays should be used to complement and confirm results generated by ELISA, which is prone to false positivity. In the evaluation of antibody-mediated virus neutralization in external secretions, precautions and rigorous controls must be used to exclude the effect of innate humoral factors. The evaluation of cell-mediated immune responses in mucosal tissues is difficult due to the low yields of cells obtained from tissue biopsies or cytobrush scrapings. Furthermore, tissue biopsies of, for example rectal mucosa, provide information pertinent exclusively to this local site, which due to the differences in distribution of cells of different phenotypes, do not provide information generalized to the entire intestinal tract. Importantly, studies concerning the kinetics of cellular responses are difficult to perform due to the limited availability of samples or to the inability of obtaining frequent repeated tissue biopsies. For sampling the female genital tract parallel collection of menstrual and peripheral blood yields high numbers of cells that permit their detailed phenotypic and functional analyses. In contrast to tissue biopsies, this non-traumatic collection procedure, results in high cell yields and repeated monthly sampling permits extensive and parallel functional studies of kinetics and unique characteristics of HIV-1-specific cellular responses in the female genital tract and peripheral blood.

12.
Kidney Blood Press Res ; 43(2): 350-359, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29529610

RESUMO

BACKGROUND/AIMS: IgA nephropathy is associated with aberrant O-glycosylation of IgA1, which is recognized by autoantibodies leading to the formation of circulating immune complexes. Some of them, after deposition into kidney mesangium, trigger glomerular injury. In patients with active disease nonresponding to angiotensin-converting enzyme inhibitors or angiotensin II blockers, corticosteroids are recommended. METHODS: The relationship between the corticosteroid therapy and serum levels of IgA, aberrantly O-glycosylated IgA1, IgA-containing immune complexes and their mesangioproliferative activity was analyzed in IgA nephropathy patients and disease and healthy controls. RESULTS: Prednisone therapy significantly reduced proteinuria and levels of serum IgA, galactose-deficient IgA1, and IgA-IgG immune complexes in IgA nephropathy patients and thus reduced differences in all of the above parameters between IgAN patients and control groups. A moderate but not significant reduction of mesangioproliferative potential of IgA-IgG immune complexes and IgA sialylation was detected. CONCLUSION: The prednisone therapy reduces overall aberrancy in IgA1 O-glycosylation in IgA nephropathy patients, but the measurement of IgA1 parameters does not allow us to predict the prednisone therapy outcome in individual patients.


Assuntos
Glomerulonefrite por IGA/tratamento farmacológico , Glucocorticoides/farmacologia , Glicosilação/efeitos dos fármacos , Imunoglobulina A/metabolismo , Anticorpos/sangue , Complexo Antígeno-Anticorpo/sangue , Estudos de Casos e Controles , Glomerulonefrite por IGA/diagnóstico , Glucocorticoides/uso terapêutico , Humanos , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Prednisona/uso terapêutico , Prognóstico
13.
PLoS One ; 12(7): e0180245, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28671952

RESUMO

Mechanisms responsible for natural control of human immunodeficiency type 1 (HIV) replication in elite controllers (EC) remain incompletely defined. To determine if EC generate high quality HIV-specific IgA responses, we used Western blotting to compare the specificities and frequencies of IgA to HIV antigens in serum of gender-, age- and race-matched EC and aviremic controllers (HC) and viremic noncontrollers (HN) on highly active antiretroviral therapy (HAART). Concentrations and avidity of IgA to HIV antigens were measured using ELISA or multiplex assays. Measurements for IgG were performed in parallel. EC were found to have stronger p24- and V1V2-specific IgG responses than HN, but there were no IgG differences for EC and HC. In contrast, IgA in EC serum bound more frequently to gp160 and gag proteins than IgA in HC or HN. The avidity of anti-gp41 IgA was also greater in EC, and these subjects had stronger IgA responses to the gp41 heptad repeat region 1 (HR1), a reported target of anti-bacterial RNA polymerase antibodies that cross react with gp41. However, EC did not demonstrate greater IgA responses to E. coli RNA polymerase or to peptides containing the shared LRAI sequence, suggesting that most of their HR1-specific IgA antibodies were not induced by intestinal microbiota. In both EC and HAART recipients, the concentrations of HIV-specific IgG were greater than HIV-specific IgA, but their avidities were comparable, implying that they could compete for antigen. Exceptions were C1 peptides and V1V2 loops. IgG and IgA responses to these antigens were discordant, with IgG reacting to V1V2, and IgA reacting to C1, especially in EC. Interestingly, EC with IgG hypergammaglobulinemia had greater HIV-specific IgA and IgG responses than EC with normal total IgG levels. Heterogeneity in EC antibody responses may therefore be due to a more focused HIV-specific B cell response in some of these individuals. Overall, these data suggest that development of HIV-specific IgA responses and affinity maturation of anti-gp41 IgA antibodies occurs to a greater extent in EC than in subjects on HAART. Future studies will be required to determine if IgA antibodies in EC may contribute in control of viral replication.


Assuntos
Terapia Antirretroviral de Alta Atividade , Proteína gp41 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Imunoglobulina A/sangue , Adulto , Afinidade de Anticorpos , Ensaio de Imunoadsorção Enzimática , Feminino , Anticorpos Anti-HIV/biossíntese , Infecções por HIV/tratamento farmacológico , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade
15.
PLoS Pathog ; 13(1): e1006087, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28125732

RESUMO

HIV-1 infection is associated with an early and profound depletion of mucosal memory CD4+ T cells, a population that plays an indispensable role in the regulation of isotype switching and transepithelial transport of antibodies. In this study, we addressed whether the depletion of CD4+ T cell in HIV-1-infected individuals results in altered humoral responses specific to antigens encountered at mucosal surfaces. Comprehensive protein microarray of systemic humoral responses to intestinal microbiota demonstrated reduced IgG responses to antigens derived from Proteobacteria and Firmicutes but not Bacteroidetes. Importantly, intestinal secretions of antiretroviral therapy-treated HIV-1-infected individuals exhibited a significant elevation of IgM levels and decreased IgA/IgM and IgG/IgM ratios of antibodies specific to a variety of microbial and food antigens. The presented findings indicate reduced competence of mucosal B cells for class switch recombination from IgM to other isotypes limiting their capacity to react to changing antigenic variety in the gut lumen. Decreased availability of microbiota-specific IgA and IgG may be an important factor contributing to the translocation of microbial antigens across the intestinal mucosal barrier and their systemic dissemination that drives chronic inflammation in HIV-1-infected individuals.


Assuntos
Antígenos/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Microbiota/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Doença Crônica , Alimentos , Regulação da Expressão Gênica , Infecções por HIV/virologia , Humanos , Imunidade Humoral , Imunoglobulina A/imunologia , Switching de Imunoglobulina , Imunoglobulina G/imunologia , Inflamação , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia
16.
Clin Immunol ; 172: 72-77, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27444044

RESUMO

IgA nephropathy (IgAN) is the leading cause of primary glomerulonephritis in the world. The disease is characterized by the presence of IgA-containing immune complexes in the circulation and in mesangial deposits with ensuing glomerular injury. Although in humans there are two IgA subclasses, only IgA1 molecules are involved. The exclusivity of participation of IgA1 in IgAN prompted extensive structural and immunological studies of the unique hinge region (HR) of IgA1, which is absent in otherwise highly homologous IgA2. HR of IgA1 with altered O-glycans serves as an antigen recognized by autoantibodies specific for aberrant HR glycans leading to the generation of nephritogenic immune complexes. However, there are several unresolved questions concerning the phylogenetic origin of human IgA1 HR, the structural basis of its antigenicity, the origin of antibodies specific for HR with altered glycan moieties, the regulatory defects in IgA1 glycosylation pathways, and the potential approaches applicable to the disease-specific interventions in the formation of nephritogenic immune complexes. This review focuses on the gaps in our knowledge of molecular and cellular events that are involved in the immunopathogenesis of IgAN.


Assuntos
Glomerulonefrite por IGA/imunologia , Animais , Modelos Animais de Doenças , Humanos , Imunoglobulina A/imunologia
17.
Front Immunol ; 7: 117, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27148252

RESUMO

IgA nephropathy (IgAN) is the most common primary glomerulonephritis, frequently leading to end-stage renal disease, as there is no disease-specific therapy. IgAN is diagnosed from pathological assessment of a renal biopsy specimen based on predominant or codominant IgA-containing immunodeposits, usually with complement C3 co-deposits and with variable presence of IgG and/or IgM. The IgA in these renal deposits is galactose-deficient IgA1, with less than a full complement of galactose residues on the O-glycans in the hinge region of the heavy chains. Research from the past decade led to the definition of IgAN as an autoimmune disease with a multi-hit pathogenetic process with contributing genetic and environmental components. In this process, circulating galactose-deficient IgA1 (autoantigen) is bound by antiglycan IgG or IgA (autoantibodies) to form immune complexes. Some of these circulating complexes deposit in glomeruli, and thereby activate mesangial cells and induce renal injury through cellular proliferation and overproduction of extracellular matrix components and cytokines/chemokines. Glycosylation pathways associated with production of the autoantigen and the unique characteristics of the corresponding autoantibodies in patients with IgAN have been uncovered. Complement likely plays a significant role in the formation and the nephritogenic activities of these complexes. Complement activation is mediated through the alternative and lectin pathways and probably occurs systemically on IgA1-containing circulating immune complexes as well as locally in glomeruli. Incidence of IgAN varies greatly by geographical location; the disease is rare in central Africa but accounts for up to 40% of native-kidney biopsies in eastern Asia. Some of this variation may be explained by genetically determined influences on the pathogenesis of the disease. Genome-wide association studies to date have identified several loci associated with IgAN. Some of these loci are associated with the increased prevalence of IgAN, whereas others, such as deletion of complement factor H-related genes 1 and 3, are protective against the disease. Understanding the molecular mechanisms and genetic and biochemical factors involved in formation and activities of pathogenic IgA1-containing immune complexes will enable the development of future disease-specific therapies as well as identification of non-invasive disease-specific biomarkers.

18.
Vaccine ; 34(27): 3171-3177, 2016 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-27113165

RESUMO

The prevalent human papillomaviruses (HPVs) infect human epithelial tissues. Infections by the mucosotropic HPV genotypes cause hyperproliferative ano-genital lesions. Persistent infections by high-risk (HR) HPVs such as HPV-16, HPV-18 and related types can progress to high grade intraepithelial neoplasias and cancers. Prophylactic HPV vaccines are based on DNA-free virus-like particles (VLPs) composed of the major capsid protein L1 of HPV-16, -18, -6 and -11 (Gardasil) or HPV-16 and -18 (Cervarix). Sera from vaccinated animals effectively prevent HPV pseudovirions to infect cell lines and mouse cervical epithelia. Both vaccines have proven to be highly protective in people. HPV pseudovirions are assembled in HEK293TT cells from matched L1 and L2 capsid proteins to encapsidate a reporter gene. Pseudovirions and genuine virions have structural differences and they infect cell lines or primary human keratinocytes (PHKs) with different efficiencies. In this study, we show that sera and isolated IgG from women immunized with Gardasil prevent authentic HPV-18 virions from infecting PHKs, whereas non-immune sera and purified IgG thereof are uniformly ineffective. Using early passage PHKs, neutralization is achieved only if immune sera are added within 2-4h of infection. We attribute the timing effect to a conformational change in HPV virions, thought to occur upon initial binding to heparan sulfate proteoglycans (HSPG) on the cell surface. This interpretation is consistent with the inability of immune IgG bound to or taken up by PHKs to neutralize the virus. Interestingly, the window of neutralization increases to 12-16h in slow growing, late passage PHKs, suggestive of altered cell surface molecules. In vivo, this window might be further lengthened by the time required to activate the normally quiescent basal cells to become susceptible to infection. Our observations help explain the high efficacy of HPV vaccines.


Assuntos
Anticorpos Antivirais/imunologia , Vacina Quadrivalente Recombinante contra HPV tipos 6, 11, 16, 18/uso terapêutico , Soros Imunes/imunologia , Queratinócitos/virologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Células Cultivadas , Feminino , Proteoglicanas de Heparan Sulfato/metabolismo , Papillomavirus Humano 18 , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Testes de Neutralização , Infecções por Papillomavirus/prevenção & controle
19.
Pathog Immun ; 1(2): 371-387, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28164171

RESUMO

BACKGROUND: Tissue resident memory T cells (TrM) provide an enhanced response against infection at mucosal surfaces, yet their function has not been extensively studied in humans, including the female genital tract (FGT). METHODS: Using polychromatic flow cytometry, we studied TrM cells, defined as CD62L-CCR7-CD103+CD69+ CD4+ and CD8+ T cells in mucosa-derived T cells from healthy and HIV-positive women. RESULTS: We demonstrate that TrM are present in the FGT of healthy and HIV-positive women. The expression of the mucosal retention receptor, CD103, from HIV-positive women was reduced compared to healthy women and was lowest in women with CD4 counts < 500 cells/mm3. Furthermore, CD103 expression on mucosa-derived CD8+ T cells correlated with antigen-specific IFN-γ production by mucosal CD4+ T cells and was inversely correlated with T-bet from CD8+CD103+ mucosa-derived T cells. CONCLUSIONS: These data suggest that CD4+ T cells, known to be impaired during HIV-1 infection and necessary for the expression of CD103 in murine models, may play a role in the expression of CD103 on resident T cells from the human FGT.

20.
Vnitr Lek ; 62 Suppl 6: 67-77, 2016.
Artigo em Tcheco | MEDLINE | ID: mdl-28124935

RESUMO

IgA nephropathy (IgAN) is the most common type of glomerulonephritis. Its etiology involves an increased production of polymeric immunoglobulin A1 with an abnormal composition of some carbohydrate chains. The reaction of these abnormal forms of IgA1 with specific autoantibodies while circulating immune complexes arise and settle in the renal mesangium with subsequent inflammatory activation of mesangial cells which in up to 50% of cases results in end-stage kidney failure. Pathogenesis involves an interplay of genetic predisposition and environmental effects, mainly of microbial nature. Current therapy is not sufficiently effective and lacks the focus on the cause of the disease, therefore more efficient and specific ways of therapy are being sought to target the individual stages of the pathogenetic process of IgAN development. With the accumulation of knowledge, new questions arise, concerning detailed mechanisms of the pathological processes, as discussed in the text.Key words: autoimmunity - glycosylation of IgA hinge region - IgA nephropathy - immunoglobulin IgA - IgA1 hinge region.


Assuntos
Mesângio Glomerular/fisiopatologia , Glomerulonefrite por IGA , Falência Renal Crônica , Glomerulonefrite por IGA/etiologia , Glomerulonefrite por IGA/terapia , Glicosilação , Humanos , Imunoglobulina A , Rim
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...