Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.970
Filtrar
1.
Front Immunol ; 13: 930112, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774784

RESUMEN

The risk to develop ACPA positive rheumatoid arthritis (RA), the most destructive type of autoimmune arthritis, is carried by HLA-DRB1 alleles containing a 5 amino acid motif: the shared epitope (SE). RA is preceded by the emergence of disease specific anti citrullinated protein antibodies (ACPA). SE positive HLA-DRB1 alleles are associated with ACPA and ACPA positive RA, not with ACPA negative RA, suggesting that ACPA contribute to the pathogenesis of RA. Understanding how HLA-DRB1 genotypes influence ACPA could lead to a curative or preventive treatment of RA. The "Shared epitope binds citrullinated peptides " hypothesis suggests that RA associated HLA-DR alleles present citrullinated peptides to T cells that help ACPA producing B cells. The "Hapten carrier model" suggests that PAD4 is the target of the T cells which help ACPA specific B cells through a hapten carrier mechanism in which PAD4 is the carrier and citrullinated peptides are the haptens. Direct binding assay of citrullinated peptides to purified HLA-DR molecules does not support the "shared epitope binds citrullinated peptides" hypothesis. The Odds Ratios to develop ACPA positive RA associated with each of 12 common HLA-DRB1 genotypes match the probability that the two HLA-DR molecules they encode can bind at least one peptide from PAD4, not from citrullinated fibrinogen. Thus, PAD4 tolerization might stop the carrier effect and switch off production of ACPA.


Asunto(s)
Artritis Reumatoide , Autoanticuerpos , Citrulinación , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Autoanticuerpos/biosíntesis , Epítopos , Cadenas HLA-DRB1/genética , Cadenas HLA-DRB1/inmunología , Haptenos/genética , Haptenos/inmunología , Humanos , Péptidos/genética , Péptidos/inmunología , Péptidos Cíclicos/genética , Péptidos Cíclicos/inmunología
2.
Viruses ; 14(1)2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35062304

RESUMEN

Viruses are a possible cause for Sjögren's syndrome (SS) as an environmental factor related to SS onset, which exhibits exocrine gland dysfunction and the emergence of autoantibodies. Although retroviruses may exhibit lymphocytic infiltration into exocrine glands, human T-cell leukemia virus type 1 (HTLV-1) has been postulated to be a causative agent for SS. Transgenic mice with HTLV-1 genes showed sialadenitis resembling SS, but their phenotypic symptoms differed based on the adopted region of HTLV-1 genes. The dominance of tax gene differed in labial salivary glands (LSGs) of SS patients with HTLV 1-associated myelopathy (HAM) and adult T-cell leukemia. Although HTLV-1 was transmitted to salivary gland epithelial cells (SGECs) by a biofilm-like structure, no viral synapse formation was observed. After infection to SGECs derived from SS patients, adhesion molecules and migration factors were time-dependently released from infected SGECs. The frequency of the appearance of autoantibodies including anti-Ro/SS-A, La/SS-B antibodies in SS patients complicated with HAM is unknown; the observation of less frequent ectopic germinal center formation in HTLV-1-seropositive SS patients was a breakthrough. In addition, HTLV-1 infected cells inhibited B-lymphocyte activating factor or C-X-C motif chemokine 13 through direct contact with established follicular dendritic cell-like cells. These findings show that HTLV-1 is directly involved in the pathogenesis of SS.


Asunto(s)
Infecciones por HTLV-I , Síndrome de Sjögren/virología , Animales , Autoanticuerpos/biosíntesis , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Genes Virales , Infecciones por HTLV-I/complicaciones , Infecciones por HTLV-I/epidemiología , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Humanos , Linfocitos/virología , Ratones , Ratones Transgénicos , Paraparesia Espástica Tropical/complicaciones , Paraparesia Espástica Tropical/epidemiología , Paraparesia Espástica Tropical/inmunología , Paraparesia Espástica Tropical/virología , Fenotipo , Ratas , Proteínas de los Retroviridae/genética , Proteínas de los Retroviridae/metabolismo , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Glándulas Salivales/virología , Síndrome de Sjögren/epidemiología , Síndrome de Sjögren/inmunología
3.
Blood ; 138(22): 2256-2268, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34587242

RESUMEN

SARS-CoV-2 vaccine ChAdOx1 nCoV-19 (AstraZeneca) causes a thromboembolic complication termed vaccine-induced immune thrombotic thrombocytopenia (VITT). Using biophysical techniques, mouse models, and analysis of VITT patient samples, we identified determinants of this vaccine-induced adverse reaction. Super-resolution microscopy visualized vaccine components forming antigenic complexes with platelet factor 4 (PF4) on platelet surfaces to which anti-PF4 antibodies obtained from VITT patients bound. PF4/vaccine complex formation was charge-driven and increased by addition of DNA. Proteomics identified substantial amounts of virus production-derived T-REx HEK293 proteins in the ethylenediaminetetraacetic acid (EDTA)-containing vaccine. Injected vaccine increased vascular leakage in mice, leading to systemic dissemination of vaccine components known to stimulate immune responses. Together, PF4/vaccine complex formation and the vaccine-stimulated proinflammatory milieu trigger a pronounced B-cell response that results in the formation of high-avidity anti-PF4 antibodies in VITT patients. The resulting high-titer anti-PF4 antibodies potently activated platelets in the presence of PF4 or DNA and polyphosphate polyanions. Anti-PF4 VITT patient antibodies also stimulated neutrophils to release neutrophil extracellular traps (NETs) in a platelet PF4-dependent manner. Biomarkers of procoagulant NETs were elevated in VITT patient serum, and NETs were visualized in abundance by immunohistochemistry in cerebral vein thrombi obtained from VITT patients. Together, vaccine-induced PF4/adenovirus aggregates and proinflammatory reactions stimulate pathologic anti-PF4 antibody production that drives thrombosis in VITT. The data support a 2-step mechanism underlying VITT that resembles the pathogenesis of (autoimmune) heparin-induced thrombocytopenia.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Autoanticuerpos/inmunología , COVID-19/prevención & control , Proteínas de la Cápside/efectos adversos , ChAdOx1 nCoV-19/efectos adversos , Contaminación de Medicamentos , Vectores Genéticos/efectos adversos , Células HEK293/inmunología , Inmunoglobulina G/inmunología , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/etiología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/efectos adversos , Adenoviridae/inmunología , Animales , Complejo Antígeno-Anticuerpo/ultraestructura , Autoanticuerpos/biosíntesis , Síndrome de Fuga Capilar/etiología , Proteínas de la Cápside/inmunología , Línea Celular Transformada , ChAdOx1 nCoV-19/química , ChAdOx1 nCoV-19/inmunología , ChAdOx1 nCoV-19/toxicidad , Dispersión Dinámica de Luz , Epítopos/química , Epítopos/inmunología , Trampas Extracelulares/inmunología , Extravasación de Materiales Terapéuticos y Diagnósticos/etiología , Vectores Genéticos/inmunología , Células HEK293/química , Humanos , Imagenología Tridimensional , Inmunoglobulina G/biosíntesis , Inflamación , Ratones , Microscopía/métodos , Activación Plaquetaria , Proteómica , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/inmunología , Trombosis de los Senos Intracraneales/diagnóstico por imagen , Trombosis de los Senos Intracraneales/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Cultivo de Virus
5.
Mol Cells ; 44(6): 392-400, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34059562

RESUMEN

It has been more than a year since severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first emerged. Many studies have provided insights into the various aspects of the immune response in coronavirus disease 2019 (COVID-19). Especially for antibody treatment and vaccine development, humoral immunity to SARS-CoV-2 has been studied extensively, though there is still much that is unknown and controversial. Here, we introduce key discoveries on the humoral immune responses in COVID-19, including the immune dynamics of antibody responses and correlations with disease severity, neutralizing antibodies and their cross-reactivity, how long the antibody and memory B-cell responses last, aberrant autoreactive antibodies generated in COVID-19 patients, and the efficacy of currently available therapeutic antibodies and vaccines against circulating SARS-CoV-2 variants, and highlight gaps in the current knowledge.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Linfocitos B/inmunología , COVID-19/inmunología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/biosíntesis , SARS-CoV-2/patogenicidad , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/biosíntesis , Acrecentamiento Dependiente de Anticuerpo , Autoanticuerpos/biosíntesis , Linfocitos B/virología , COVID-19/mortalidad , COVID-19/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina A/biosíntesis , Inmunoglobulina M/biosíntesis , Memoria Inmunológica , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Tratamiento Farmacológico de COVID-19
6.
Clin Appl Thromb Hemost ; 27: 10760296211021498, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34060379

RESUMEN

Today the coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has become a global health problem. After more than a year with the pandemic, although our knowledge has progressed on COVID-19, there are still many unknowns in virological, pathophysiological and immunological aspects. It is obvious that the most efficient solution to end this pandemic are safe and efficient vaccines. This manuscript summarizes the pathophysiological and thrombotic features of COVID-19 and the safety and efficacy of currently approved COVID-19 vaccines with an aim to clarify the recent concerns of thromboembolic events after COVID-19 vaccination. The influx of newer information is rapid, requiring periodic updates and objective assessment of the data on the pathogenesis of COVID-19 variants and the safety and efficacy of currently available vaccines.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , SARS-CoV-2 , Trombosis/etiología , Ad26COVS1 , Autoanticuerpos/biosíntesis , Vacuna BNT162 , COVID-19/epidemiología , COVID-19/fisiopatología , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , ChAdOx1 nCoV-19 , Ensayos Clínicos como Asunto , Coagulación Intravascular Diseminada/epidemiología , Coagulación Intravascular Diseminada/etiología , Aprobación de Drogas , Femenino , Vectores Genéticos , Glicosaminoglicanos/inmunología , Humanos , Masculino , Modelos Cardiovasculares , Pandemias/prevención & control , Factor Plaquetario 4/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Seguridad , Trombosis de los Senos Intracraneales/epidemiología , Trombosis de los Senos Intracraneales/etiología , Trombosis/epidemiología , Trombosis/fisiopatología , Vacunas de Productos Inactivados/efectos adversos , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de ARNm
7.
Front Immunol ; 12: 676046, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054864

RESUMEN

Non-infectious uveitis is an inflammatory disorder of the eye that accounts for severe visual loss without evident infectious agents. While T cells are supposed to dominate the induction of inflammation in non-infectious uveitis, the role of B cells in the pathogenesis of this disease is obscure. Therefore, this review aimed to discuss diverse B-cell participation in different non-infectious uveitides and their roles in the pathogenesis of this disease as well as the mechanism of action of rituximab. Increasing evidence from experimental models and human non-infectious uveitis has suggested the participation of B cells in non-infectious uveitis. The participation levels vary in different uveitides. Furthermore, B cells play multiple roles in the pathogenic mechanisms. B cells produce autoantibodies, regulate T cell responses via antibody-independent functions, and constitute ectopic lymphoid structures. Regulatory B cells perform pivotal anti-inflammatory functions in non-infectious uveitis. Rituximab may work by depleting pro-inflammatory B cells and restoring the quantity and function of regulatory B cells in this disease. Identifying the levels of B-cell participation and the associated roles is beneficial for optimizing therapy. Diversified experimental model choices and emerging tools and/or methods are conducive for future studies on this topic.


Asunto(s)
Linfocitos B/fisiología , Uveítis/inmunología , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/etiología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Humanos , Rituximab/uso terapéutico , Uveítis/etiología
8.
Front Immunol ; 12: 676082, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959133

RESUMEN

Lupus nephritis (LN) is a common complication of systemic lupus erythematosus (SLE) and a major risk factor for morbidity and mortality. The abundant cell-free nucleic (DNA/RNA) in SLE patients, especially dsDNA, is a key substance in the pathogenesis of SLE and LN. The deposition of DNA/RNA-immune complexes (DNA/RNA-ICs) in the glomerulus causes a series of inflammatory reactions that lead to resident renal cell disturbance and eventually renal fibrosis. Cell-free DNA/RNA is the most effective inducer of type I interferons (IFN-I). Resident renal cells (rather than infiltrating immune cells) are the main source of IFN-I in the kidney. IFN-I in turn damages resident renal cells. Not only are resident renal cells victims, but also participants in this immunity war. However, the mechanism for generation of IFN-I in resident renal cells and the pathological mechanism of IFN-I promoting renal fibrosis have not been fully elucidated. This paper reviews the latest epidemiology of LN and its development process, discusses the mechanism for generation of IFN-I in resident renal cells and the role of IFN-I in the pathogenesis of LN, and may open a new perspective for the treatment of LN.


Asunto(s)
Interferón Tipo I/fisiología , Riñón/patología , Nefritis Lúpica/etiología , Complejo Antígeno-Anticuerpo/metabolismo , Autoanticuerpos/biosíntesis , Fibrosis , Humanos , Riñón/irrigación sanguínea , Riñón/inmunología , Glomérulos Renales/patología
9.
Blood ; 137(26): 3656-3659, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33945605

RESUMEN

Vaccination is crucial in combatting the severe acute respiratory syndrome coronavirus 2 pandemic. The rare complication of thrombocytopenia and thrombotic complications at unusual sites after ChAdOx1 nCov-19 vaccination is caused by platelet-activating antibodies directed against platelet factor 4 (PF4). We present a widely applicable whole-blood standard flow cytometric assay to identify the pathogenic antibodies associated with vaccine-induced immune-mediated thrombotic thrombocytopenia (VITT) after ChAdOx1 nCov-19 vaccination. This assay will enable rapid diagnosis by many laboratories. This trial was registered at www.clinicaltrials.gov as #NCT04370119.


Asunto(s)
Autoanticuerpos/sangre , Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , Citometría de Flujo/métodos , Inmunoglobulina G/sangre , Activación Plaquetaria/inmunología , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/diagnóstico , Receptores de IgG/inmunología , SARS-CoV-2 , Vacunación/efectos adversos , Especificidad de Anticuerpos , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Vacunas contra la COVID-19/inmunología , ChAdOx1 nCoV-19 , Heparina/efectos adversos , Heparina/inmunología , Humanos , Técnicas para Inmunoenzimas , Inmunogenicidad Vacunal , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Selectina-P/análisis , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/inmunología
10.
Biochem J ; 478(8): 1485-1509, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33881487

RESUMEN

Carbohydrate-binding antibodies play diverse and critical roles in human health. Endogenous carbohydrate-binding antibodies that recognize bacterial, fungal, and other microbial carbohydrates prevent systemic infections and help maintain microbiome homeostasis. Anti-glycan antibodies can have both beneficial and detrimental effects. For example, alloantibodies to ABO blood group carbohydrates can help reduce the spread of some infectious diseases, but they also impose limitations for blood transfusions. Antibodies that recognize self-glycans can contribute to autoimmune diseases, such as Guillain-Barre syndrome. In addition to endogenous antibodies that arise through natural processes, a variety of vaccines induce anti-glycan antibodies as a primary mechanism of protection. Some examples of approved carbohydrate-based vaccines that have had a major impact on human health are against pneumococcus, Haemophilus influeanza type b, and Neisseria meningitidis. Monoclonal antibodies specifically targeting pathogen associated or tumor associated carbohydrate antigens (TACAs) are used clinically for both diagnostic and therapeutic purposes. This review aims to highlight some of the well-studied and critically important applications of anti-carbohydrate antibodies.


Asunto(s)
Síndrome de Guillain-Barré/inmunología , Infecciones por Haemophilus/inmunología , Meningitis Meningocócica/inmunología , Neumonía Neumocócica/inmunología , Polisacáridos/inmunología , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Vacunas Bacterianas/biosíntesis , Vacunas Bacterianas/uso terapéutico , Secuencia de Carbohidratos , Síndrome de Guillain-Barré/patología , Infecciones por Haemophilus/microbiología , Infecciones por Haemophilus/prevención & control , Vacunas contra Haemophilus/biosíntesis , Vacunas contra Haemophilus/uso terapéutico , Haemophilus influenzae/inmunología , Humanos , Meningitis Meningocócica/microbiología , Meningitis Meningocócica/prevención & control , Neisseria meningitidis/inmunología , Vacunas Neumococicas/biosíntesis , Vacunas Neumococicas/uso terapéutico , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/prevención & control , Polisacáridos/antagonistas & inhibidores , Polisacáridos/química , Streptococcus pneumoniae/inmunología
11.
Science ; 371(6534)2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33707237

RESUMEN

Antiphospholipid antibodies (aPLs) cause severe autoimmune disease characterized by vascular pathologies and pregnancy complications. Here, we identify endosomal lysobisphosphatidic acid (LBPA) presented by the CD1d-like endothelial protein C receptor (EPCR) as a pathogenic cell surface antigen recognized by aPLs for induction of thrombosis and endosomal inflammatory signaling. The engagement of aPLs with EPCR-LBPA expressed on innate immune cells sustains interferon- and toll-like receptor 7-dependent B1a cell expansion and autoantibody production. Specific pharmacological interruption of EPCR-LBPA signaling attenuates major aPL-elicited pathologies and the development of autoimmunity in a mouse model of systemic lupus erythematosus. Thus, aPLs recognize a single cell surface lipid-protein receptor complex to perpetuate a self-amplifying autoimmune signaling loop dependent on the cooperation with the innate immune complement and coagulation pathways.


Asunto(s)
Presentación de Antígeno , Autoinmunidad , Coagulación Sanguínea/inmunología , Receptor de Proteína C Endotelial/inmunología , Lupus Eritematoso Sistémico/inmunología , Lisofosfolípidos/inmunología , Monoglicéridos/inmunología , Animales , Anticuerpos Antifosfolípidos/biosíntesis , Autoanticuerpos/biosíntesis , Modelos Animales de Enfermedad , Pérdida del Embrión/inmunología , Endosomas/inmunología , Receptor de Proteína C Endotelial/genética , Humanos , Inmunidad Innata , Lupus Eritematoso Sistémico/sangre , Ratones , Ratones Mutantes , Esfingomielina Fosfodiesterasa/metabolismo , Trombosis/inmunología , Receptor Toll-Like 7/inmunología
12.
Nat Commun ; 12(1): 1032, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589587

RESUMEN

Pulmonary alveolar proteinosis (PAP) is a devastating lung disease caused by abnormal surfactant homeostasis, with a prevalence of 6-7 cases per million population worldwide. While mutations causing hereditary PAP have been reported, the genetic basis contributing to autoimmune PAP (aPAP) has not been thoroughly investigated. Here, we conducted a genome-wide association study of aPAP in 198 patients and 395 control participants of Japanese ancestry. The common genetic variant, rs138024423 at 6p21, in the major-histocompatibility-complex (MHC) region was significantly associated with disease risk (Odds ratio [OR] = 5.2; P = 2.4 × 10-12). HLA fine-mapping revealed that the common HLA class II allele, HLA-DRB1*08:03, strongly drove this signal (OR = 4.8; P = 4.8 × 10-12), followed by an additional independent risk allele at HLA-DPß1 amino acid position 8 (OR = 0.28; P = 3.4 × 10-7). HLA-DRB1*08:03 was also associated with an increased level of anti-GM-CSF antibody, a key driver of the disease (ß = 0.32; P = 0.035). Our study demonstrated a heritable component of aPAP, suggesting an underlying genetic predisposition toward an abnormal antibody production.


Asunto(s)
Autoanticuerpos/genética , Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Cadenas HLA-DRB1/genética , Proteinosis Alveolar Pulmonar/genética , Adulto , Anciano , Alelos , Pueblo Asiatico , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/etnología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Estudios de Casos y Controles , Cromosomas Humanos Par 6 , Femenino , Expresión Génica , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Cadenas HLA-DRB1/inmunología , Humanos , Japón , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Isoformas de Proteínas/genética , Proteinosis Alveolar Pulmonar/etnología , Proteinosis Alveolar Pulmonar/inmunología , Proteinosis Alveolar Pulmonar/patología , Surfactantes Pulmonares/inmunología , Surfactantes Pulmonares/metabolismo , Riesgo
13.
Clin Appl Thromb Hemost ; 27: 1076029620977702, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539214

RESUMEN

The SARS-CoV-2 pandemic has focused attention on prevention, restriction and treatment methods that are acceptable worldwide. This means that they should be simple and inexpensive. This review examines the possible role of glycosaminoglycan (GAG) antithrombotics in the treatment of COVID-19. The pathophysiology of this disease reveals a complex interplay between the hemostatic and immune systems that can be readily disrupted by SARS-CoV-2. Some of the GAG antithrombotics also possess immune-modulatory actions and since they are relatively inexpensive they could play an important role in the management of COVID-19 and its complications.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Fibrinolíticos/uso terapéutico , Heparina/uso terapéutico , Autoanticuerpos/biosíntesis , COVID-19/complicaciones , COVID-19/fisiopatología , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Endotelio Vascular/virología , Femenino , Glicosaminoglicanos/uso terapéutico , Hemorragia/etiología , Trastornos Hemostáticos/tratamiento farmacológico , Trastornos Hemostáticos/etiología , Trastornos Hemostáticos/fisiopatología , Humanos , Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/fisiopatología , Masculino , Pandemias , Factores de Riesgo , SARS-CoV-2/patogenicidad , Trombina/biosíntesis , Trombosis/etiología
14.
Blood Coagul Fibrinolysis ; 32(4): 294-297, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33443928

RESUMEN

Factor V inhibitors are a rare cause of life-threatening bleeding. We present a case of an acquired factor V inhibitor likely caused by coronavirus disease 2019 infection. Bleeding was manifested by severe anemia requiring frequent red-cell transfusion, left psoas muscle hematoma, and left retroperitoneal cavity hematoma. Factor V activity was less than 1% and the factor V inhibitor titer was 31.6 Bethesda units. Severe acute respiratory syndrome coronavirus 2 RNA testing of the nasopharynx was positive 2 weeks before presentation and continued to be positive for 30 days. The patient failed treatment with intravenous immunoglobulin and dexamethasone. Three cycles of plasmapheresis with fresh frozen plasma replacement resulted in correction of the bleeding and laboratory coagulopathy. This is the first reported case of a factor V inhibitor in a coronavirus disease 2019 patient and suggests that plasmapheresis may be a successful treatment strategy.


Asunto(s)
Autoanticuerpos/biosíntesis , COVID-19/sangre , Factor V/inmunología , Trastornos Hemorrágicos/etiología , SARS-CoV-2 , Anciano de 80 o más Años , Anemia/etiología , Anemia/terapia , Anticuerpos Antivirales/sangre , Especificidad de Anticuerpos , Autoanticuerpos/inmunología , COVID-19/complicaciones , COVID-19/diagnóstico , COVID-19/inmunología , Terapia Combinada , Comorbilidad , Diagnóstico Tardío , Dexametasona/uso terapéutico , Transfusión de Eritrocitos , Factor V/antagonistas & inhibidores , Femenino , Hematoma/etiología , Trastornos Hemorrágicos/tratamiento farmacológico , Trastornos Hemorrágicos/terapia , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Inhibidor de Coagulación del Lupus/sangre , Octreótido/uso terapéutico , Plasma , Plasmaféresis , SARS-CoV-2/inmunología , Vitamina K/uso terapéutico
15.
Arthritis Rheumatol ; 73(3): 478-489, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33512094

RESUMEN

OBJECTIVE: To assess the role of STAT4 activation in driving pathogenic follicular helper T (Tfh) cell secretion of the cytokines interleukin-21 (IL-21) and interferon-γ (IFNγ) in murine and human lupus. METHODS: The effect of STAT4-dependent Tfh cell signaling on cytokine production and autoreactive B cell maturation was assessed temporally during the course of lupus in a murine model, with further assessment of Tfh cell gene transcription performed using RNA-Seq technology. STAT4-dependent signaling and cytokine production were also determined in circulating Tfh-like cells in patients with systemic lupus erythematosus (SLE), as compared to cells from healthy control subjects, and correlations with disease activity were assessed in the Tfh-like cells from SLE patients. RESULTS: IL-21- and IFNγ-coproducing Tfh cells expanded prior to the detection of potentially pathogenic IgG2c autoantibodies in lupus-prone mice. Tfh cells transcriptionally evolved during the course of disease with acquisition of a STAT4-dependent gene signature. Maintenance of Tfh cell cytokine synthesis was dependent upon STAT4 signaling, driven by type I IFNs. Circulating Tfh-like cells from patients with SLE also secreted IL-21 and IFNγ, with STAT4 phosphorylation enhanced by IFNß, in association with the extent of clinical disease activity. CONCLUSION: We identified a role for type I IFN signaling in driving STAT4 activation and production of IL-21 and IFNγ by Tfh cells in murine and human lupus. Enhanced STAT4 activation in Tfh cells may underlie pathogenic B cell responses in both murine and human lupus. These data indicate that STAT4 guides pathogenic cytokine and immunoglobulin production in SLE, demonstrating a potential therapeutic target to modulate autoimmunity.


Asunto(s)
Autoanticuerpos/inmunología , Citocinas/inmunología , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/inmunología , Factor de Transcripción STAT4/inmunología , Células T Auxiliares Foliculares/inmunología , Adulto , Animales , Formación de Anticuerpos/inmunología , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulinas , Interferón gamma/inmunología , Interleucinas/inmunología , Masculino , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , RNA-Seq
16.
Dig Dis ; 39(3): 234-242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32759604

RESUMEN

BACKGROUND: To make an accurate estimate of the response to thrombopoietin (TPO) receptor agonists for thrombocytopenia associated with chronic liver disease, we evaluated the influence of antiplatelet autoantibodies on the response to lusutrombopag in thrombocytopenic patients with liver disease. METHODS: A prospective study was conducted at 2 hospitals. Thrombocytopenic patients with liver disease received oral lusutrombopag 3.0 mg once daily for up to 7 days. We analyzed changes in platelet counts from baseline to the maximum platelet count on days 9-14. The definition of clinical response was a platelet count of ≥5 × 104/µL with an increased platelet count of ≥2 × 104/µL from baseline. We assessed the correlation between the response to treatment drug and antiplatelet autoantibodies measured by anti-GPIIb/IIIa antibody-producing B cells. RESULTS: Thirty patients received the trial drug. There were 25 responders and 5 nonresponders. The median change in platelet counts was 3.9 × 104/µL (95% CI 2.8-4.6, p < 0.0001). The correlation between change in platelet counts and the frequency of the anti-glycoprotein IIb/IIIa antibody-producing B cells was moderate (r = 0.414, 95% CI 0.064-0.674, p = 0.023). In multivariate analysis of factors affecting the change in platelet counts, the anti-GPIIb/IIIa antibody-producing B cells were identified as an independent factor (regression coefficient [B] = 0.089; CI 0.021-0.157, p = 0.013). CONCLUSION: Anti-GPIIb/IIIa antibody-producing B cells may be a predictor for TPO receptor agonists in patients with chronic liver disease.


Asunto(s)
Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Cinamatos/uso terapéutico , Hepatopatías/complicaciones , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Tiazoles/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/inmunología , Anciano , Anciano de 80 o más Años , Autoanticuerpos/inmunología , Plaquetas/patología , Cinamatos/administración & dosificación , Femenino , Humanos , Hepatopatías/sangre , Masculino , Persona de Mediana Edad , Análisis Multivariante , Tamaño de los Órganos , Recuento de Plaquetas , Estudios Prospectivos , Bazo/patología , Tiazoles/administración & dosificación , Trombocitopenia/sangre , Trombocitopenia/complicaciones
17.
Methods Mol Biol ; 2225: 241-255, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33108667

RESUMEN

Systemic lupus erythematosus (SLE) is a multifactorial and heterogeneous autoimmune disease involving multiple organ systems and tissues. Lupus nephritis occurs in approximately 60% of patients with SLE and is the leading cause of morbidity. Diffuse alveolar hemorrhage (DAH) is a rare but very serious complication of SLE with a greater than 50% associated mortality. The etiology of SLE is unclear but has proposed genetic, hormonal, and environmental aspects. Pristane is a saturated terpenoid alkane and has become the most popular laboratory model for inducing lupus in mice. The pristane model of SLE has the capacity to reproduce many components of the human presentation of the disease. Previous studies have demonstrated that virus-derived immune-modulating proteins have the potential to control inflammatory and autoimmune disorders. Serp-1, a 55 kDa secreted and highly glycosylated immune modulator derived from myxoma virus (MYXV), has potent immunomodulatory activity in models of vasculitis, viral sepsis, collagen-induced arthritis, and transplant rejection. This chapter describes the mouse preclinical pristane lupus model as a method to examine virus-derived protein efficacy for treating autoimmune diseases and specifically lupus nephritis and DAH.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Hemorragia/prevención & control , Factores Inmunológicos/farmacología , Nefritis Lúpica/tratamiento farmacológico , Myxoma virus/química , Proteinuria/tratamiento farmacológico , Proteínas Virales/farmacología , Animales , Autoanticuerpos/biosíntesis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Hemorragia/inmunología , Hemorragia/patología , Humanos , Factores Inmunológicos/inmunología , Inyecciones Intraperitoneales , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Nefritis Lúpica/inducido químicamente , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Ratones , Ratones Endogámicos BALB C , Proteinuria/inducido químicamente , Proteinuria/inmunología , Proteinuria/patología , Terpenos/administración & dosificación , Resultado del Tratamiento , Proteínas Virales/inmunología
18.
Front Immunol ; 11: 581385, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33262760

RESUMEN

Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/inmunología , Células Progenitoras Endoteliales/inmunología , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Aterosclerosis/fisiopatología , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Citocinas/inmunología , Células Progenitoras Endoteliales/clasificación , Células Progenitoras Endoteliales/fisiología , Trampas Extracelulares/inmunología , Humanos , Lupus Eritematoso Sistémico/fisiopatología , Modelos Inmunológicos , Estrés Oxidativo , Transducción de Señal/inmunología , Linfocitos T/inmunología
19.
Bull Exp Biol Med ; 169(4): 431-434, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32889565

RESUMEN

We studied the effect of single and repeated intranasal administration of antibodies to glutamate in experimental parkinsonian syndrome induced by injections of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to C57BL/6J mice. Intranasal administration of anti-glutamate antibodies to mice in parallel with administration of MPTP over 10 days alleviated parkinsonian symptoms (oligokinesia and rigidity). In the serum of mice injected with antibodies to glutamate and/or MPTP, the titers of autoantibodies to glutamate and dopamine were higher than in control animals receiving saline. Single intranasal administration of anti-glutamate antibodies to mice with established parkinsonian syndrome did not affect the severity of parkinsonian symptoms.


Asunto(s)
Anticuerpos/farmacología , Antiparkinsonianos/farmacología , Dopamina/inmunología , Ácido Glutámico/inmunología , Hipocinesia/tratamiento farmacológico , Trastornos Parkinsonianos/tratamiento farmacológico , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/administración & dosificación , Administración Intranasal , Animales , Anticuerpos/química , Anticuerpos/aislamiento & purificación , Antiparkinsonianos/química , Antiparkinsonianos/aislamiento & purificación , Autoanticuerpos/biosíntesis , Dopamina/química , Ácido Glutámico/química , Caballos , Hipocinesia/inducido químicamente , Hipocinesia/inmunología , Hipocinesia/fisiopatología , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/fisiopatología , Conejos , gammaglobulinas/química , gammaglobulinas/inmunología
20.
Immunohorizons ; 4(8): 455-463, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32759326

RESUMEN

Atherosclerosis is responsible for a large percentage of all-cause mortality worldwide, but it is only now beginning to be understood as a complex disease process involving metabolic insult, chronic inflammation, and multiple immune mechanisms. Abs targeting apolipoprotein A-I (ApoA-I) have been found in patients with cardiovascular disease, autoimmune conditions, as well as those with no documented history of either. However, relatively little is known about how these Abs are generated and their relationship to diet and sex. In the current study, we modeled this aspect of autoimmunity using anti-ApoA-I immunization of male and female C57BL/6 mice. Unexpectedly, we found that autoantibodies directed against a single, previously unknown, epitope within the ApoA-I protein developed irrespective of immunization status or dyslipidemia in mice. When total IgG subclasses were analyzed over the course of time, we observed that rather than driving an increase in inflammatory IgG subclasses, consumption of Western diet suppressed age-dependent increases in IgG2b and IgG2c in male mice only. The lack of change observed in female mice suggested that diet and sex might play a combined role in Th1/Th2 balance and, ultimately, in immunity to pathogen challenge. This report demonstrates the need for inclusion of both sexes in studies pertaining to diet and aging and suggests that further study of immunogenic epitopes present in ApoA-I is warranted.


Asunto(s)
Apolipoproteína A-I/inmunología , Aterosclerosis/inmunología , Autoanticuerpos/sangre , Epítopos/inmunología , Animales , Apolipoproteína A-I/metabolismo , Autoanticuerpos/biosíntesis , Autoinmunidad , Dieta , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...