Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.341
Filtrar
Más filtros











Intervalo de año de publicación
1.
Autoimmun Rev ; 23(7-8): 103603, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39154740

RESUMEN

A growing body of evidence supports an association between systemic autoimmune disease and exposure to amphibole asbestos, a form of asbestos typically with straight, stiff, needle-like fibers that are easily inhaled. While the bulk of this evidence comes from the population exposed occupationally and environmentally to Libby Amphibole (LA) due to the mining of contaminated vermiculite in Montana, studies from Italy and Australia are broadening the evidence to other sites of amphibole exposures. What these investigations have done, that most historical studies have not, is to evaluate amphibole asbestos separately from chrysotile, the most common commercial asbestos in the United States. Here we review the current and historical evidence summarizing amphibole asbestos exposure as a risk factor for autoimmune disease. In both mice and humans, amphibole asbestos, but not chrysotile, drives production of both antinuclear autoantibodies (ANA) associated with lupus-like pathologies and pathogenic autoantibodies against mesothelial cells that appear to contribute to a severe and progressive pleural fibrosis. A growing public health concern has emerged with revelations that a) unregulated asbestos minerals can be just as pathogenic as commercial (regulated) asbestos, and b) bedrock and soil occurrences of asbestos are far more widespread than previously thought. While occupational exposures may be decreasing, environmental exposures are on the rise for many reasons, including those due to the creation of windborne asbestos-containing dusts from urban development and climate change, making this topic an urgent challenge for public and heath provider education, health screening and environmental regulations.


Asunto(s)
Asbestos Anfíboles , Enfermedades Autoinmunes , Exposición a Riesgos Ambientales , Humanos , Asbestos Anfíboles/efectos adversos , Asbestos Anfíboles/toxicidad , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/inducido químicamente , Exposición a Riesgos Ambientales/efectos adversos , Ratones , Exposición Profesional/efectos adversos
2.
Autoimmun Rev ; 23(9): 103605, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39182594

RESUMEN

Immune thrombocytopenia (ITP) is a complex autoimmune disorder characterized by thrombocytopenia and an increased bleeding risk, arising from autoantibody-mediated platelet destruction and impaired megakaryocyte function. The pathogenesis of ITP involves a multifaceted interplay of genetic predispositions, immune dysregulation, and environmental triggers, though the precise mechanisms remain uncertain. Several infectious agents, mostly viruses, have been implicated in both acute and chronic ITP through mechanisms such as molecular mimicry, direct bone marrow suppression, and immune dysregulation. Vaccinations, particularly those containing adjuvants like aluminum and those capable of inducing molecular mimicry, have also been associated with ITP, either as a new onset or as a relapse in preexisting cases. The role of drugs, particularly quinine, quinidine and certain antibiotics, in inducing ITP through various immunological pathways further illustrates the diverse etiologies of this condition. The multiple triggers of the disease raise the question of whether ITP may be classified as an autoimmune/inflammatory syndrome induced by adjuvants (ASIA). This condition encompasses a range of autoimmune and inflammatory symptoms triggered by adjuvants, such as silicones, polypropylene meshes, metal implants, and mineral oils present in various medical materials and medications. Similar to that observed in some cases of ITP, adjuvants can trigger autoimmune or autoinflammatory responses via molecular mimicry, epitope spreading, and polyclonal activation. This narrative review explores the underlying environmental factors related to ITP and examines ITP triggers that could potentially support an association between ITP and ASIA syndrome.


Asunto(s)
Adyuvantes Inmunológicos , Púrpura Trombocitopénica Idiopática , Humanos , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/etiología , Púrpura Trombocitopénica Idiopática/inducido químicamente , Adyuvantes Inmunológicos/efectos adversos , Inflamación/inmunología , Inflamación/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/etiología , Imitación Molecular/inmunología , Síndrome , Autoinmunidad
3.
RMD Open ; 10(3)2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39004431

RESUMEN

OBJECTIVES: To evaluate the long-term effectiveness and safety of autologous haematopoiesis stem cell transplantation (AHSCT) for severe, refractory autoimmune diseases in paediatric patients. METHODS: A single-centre study of consecutive children and adolescents with refractory autoimmune diseases undergoing AHSCT was performed. Demographics, clinical, laboratory features, pre-AHSCT medications, disease activity and functional status were captured. The primary outcome was progression-free survival, secondary outcomes included overall survival, disease-specific treatment responses, disease activity at the last follow-up and AHSCT safety. RESULTS: The study included seven patients: two systemic sclerosis, one pansclerotic morphoea, one eosinophilic fasciitis, one juvenile dermatomyositis and two patients with systemic juvenile idiopathic arthritis; four women, three men median age at AHSCT of 10 years (7-19), median follow-up post-AHSCT of 17 years. Median progression-free survival and overall survival was 4.2 years (95% CI: 0.98 to 8.3) and 17 years (95% CI: 11.8 to 22.1), respectively. Progression-free survival rates at 1 and 2 years post-AHSCT were 100% and 77%, respectively. All children survived. All patients are in clinical remission, only four require ongoing immunotherapy. SAFETY: Three experienced infections, including HHV6, Candida and Ralstonia sepsis; one developed a systemic inflammatory response syndrome; two new onset secondary autoimmune diseases including autoimmune haemolytic anaemia, Graves' disease and one was found to have a breast fibroadenoma. Treatment toxicity: one cyclophosphamide-associated transient renal failure and pericardial effusion, one patient with amenorrhoea/infertility. CONCLUSIONS: AHSCT was an effective and safe approach for children and adolescents with treatment-refractory autoimmune diseases. The indication and timing of transplantation requires a careful consideration and a multidisciplinary approach.


Asunto(s)
Enfermedades Autoinmunes , Trasplante de Células Madre Hematopoyéticas , Trasplante Autólogo , Humanos , Femenino , Masculino , Adolescente , Niño , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/etiología , Resultado del Tratamiento , Adulto Joven , Estudios de Seguimiento
4.
J Clin Immunol ; 44(7): 156, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38954150

RESUMEN

BACKGROUND: Thymoma presents with several autoimmune manifestations and is associated with secondary autoimmune regulator (AIRE) deficiency. Pneumonitis has recently been described as an autoimmune manifestation associated with thymoma presenting with similar clinical, radiographic, histological, and autoantibody features as seen in patients with inherited AIRE deficiency who suffer from Autoimmune PolyEndocrinopathy-Candidiasis-Ectodermal Dystrophy (APECED) syndrome. OBJECTIVES: To treat two patients with biopsy-proven thymoma-associated pneumonitis with lymphocyte-directed immunomodulation. METHODS: Two patients with thymoma were enrolled on IRB-approved protocols at the NIH Clinical Center. We performed history and physical examination; laboratory, radiographic, histologic and pulmonary function evaluations; and measurement of the lung-directed autoantibodies KCNRG and BPIFB1 prior to and at 1- and 6-months following initiation of lymphocyte-directed immunomodulation with azathioprine with or without rituximab. RESULTS: Combination T- and B-lymphocyte-directed immunomodulation resulted in improvement of clinical, functional, and radiographic parameters at 6-month follow-up evaluations in both patients with sustained remission up to 12-36 months following treatment initiation. CONCLUSION: Lymphocyte-directed immunomodulation remitted autoimmune pneumonitis in two patients with thymoma.


Asunto(s)
Inmunomodulación , Timoma , Humanos , Timoma/inmunología , Timoma/complicaciones , Timoma/diagnóstico , Femenino , Masculino , Rituximab/uso terapéutico , Autoanticuerpos/inmunología , Persona de Mediana Edad , Neoplasias del Timo/inmunología , Neoplasias del Timo/complicaciones , Neoplasias del Timo/diagnóstico , Neumonía/etiología , Neumonía/inmunología , Neumonía/diagnóstico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/etiología , Adulto , Azatioprina/uso terapéutico , Linfocitos B/inmunología , Resultado del Tratamiento , Linfocitos T/inmunología
5.
Front Immunol ; 15: 1366101, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38707905

RESUMEN

We report here the case of a 50-year-old man who was first diagnosed with myelodysplastic syndrome with excess blasts-2 (MDS-EB-2) and underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) in 2019, resulting in complete remission. However, he was diagnosed in 2021 with several autoimmune disorders, including autoimmune hepatitis (AIH), Hashimoto's thyroiditis (HT), and autoimmune hemolytic anemia (AIHA). This is referred as multiple autoimmune syndrome (MAS), which is a rare occurrence after allo-HSCT, as previously noted in the literature. Despite being treated with glucocorticoids, cyclosporine A, and other medications, the patient did not fully recover. To address the glucocorticoid-refractory MAS, a four-week course of rituximab (RTX) at a weekly dose of 100mg was administered, which significantly improved the patient's condition. Thus, this case report underscores the importance of implementing alternative treatments in patients with post-transplant autoimmune diseases, who are glucocorticoid-refractory or glucocorticoid-dependent, and highlights the effectiveness of RTX as second-line therapy.


Asunto(s)
Enfermedades Autoinmunes , Glucocorticoides , Trasplante de Células Madre Hematopoyéticas , Trasplante Homólogo , Humanos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Masculino , Persona de Mediana Edad , Glucocorticoides/uso terapéutico , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/terapia , Rituximab/uso terapéutico , Anemia Hemolítica Autoinmune/etiología , Anemia Hemolítica Autoinmune/terapia , Anemia Hemolítica Autoinmune/tratamiento farmacológico , Resistencia a Medicamentos
6.
Vaccine ; 42(18): 3830-3837, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38729910

RESUMEN

BACKGROUND: A large vaccination campaign was initiated worldwide in December 2020 in order to prevent infection with SARS-CoV-2 and severe Covid-19 disease. However, long-term adverse effects of vaccination remain unclear. Therefore, our objective was to examine the association between vaccination and the incidence of autoimmune diagnoses in the first year after vaccine uptake. METHODS: This retrospective cohort study based on Clalit Health Services (CHS) comprehensive database compared the rates of immune-mediated diagnoses among BNT162b2 vaccinated versus unvaccinated individuals. As a reference, a secondary cohort compared individuals infected with Sars-CoV-2 versus uninfected individuals. The minimum follow-up period was 4 months. The cohorts were divided into 4 age groups (12-17, 18-44, 45-64, 65 years or older). Multivariate Cox proportional hazard regression models were applied, followed by a correction for multiple comparisons using the False Discovery Rate (FDR) method, hence accounting for the investigation of multiple clinical outcomes. RESULTS: Increased risk for immune-mediated diagnoses following vaccination with BNT162b2 was observed for psoriasis in all age groups (HR 1.41-1.69), colitis among patients younger than 65 years (HR 1.38-1.93), vitiligo in patients aged 45-64 (HR 2.82, 95 %CI: 1.57-5.08) and for polymyalgia-rheumatica in patients aged 65 years or older (HR 2.12, 95 % CI: 1.3-3.47). In the reference cohort, patients who were infected by Covid-19 were at increased risk for fibromyalgia (HR 1.72, 95 % CI: 1.36-2.19 in individuals aged 18-44; HR 1.71, 95 % CI: 1.31-2.22 in individuals aged 45-64), and hypothyroidism (HR 1.54, 95 % CI: 1.15-2.07 in individuals aged 65 years or older). CONCLUSIONS: The BNT162b2 vaccine was associated with increased risk (though rare) for psoriasis, colitis and polymyalgia rheumatica. These findings should be considered as a part of the risk-benefit assessment when planning future vaccination programs for various population groups.


Asunto(s)
Vacuna BNT162 , COVID-19 , Humanos , Vacuna BNT162/inmunología , Vacuna BNT162/efectos adversos , Persona de Mediana Edad , Estudios Retrospectivos , Masculino , Adulto , Femenino , COVID-19/prevención & control , COVID-19/epidemiología , COVID-19/inmunología , Anciano , Adolescente , Adulto Joven , Incidencia , Niño , Vacunación/efectos adversos , Vacunación/estadística & datos numéricos , SARS-CoV-2/inmunología , Comorbilidad , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/etiología , Vacunas contra la COVID-19/efectos adversos , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Psoriasis/epidemiología , Psoriasis/inmunología
7.
Immunol Lett ; 268: 106869, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38788802

RESUMEN

Inflammatory bowel disease (IBD) is a chronic and progressive inflammatory intestinal disease that affects people around the world. The primary cause of IBD is an imbalance in the host immune response to intestinal flora. Several human genes, including IL10, STAT3, IRGM, ATG16L1, NOD2 and RUNX3, are associated with inappropriate immune responses in IBD. It has been reported that homozygous Runx3-knockout (ko) mice spontaneously develop colitis. However, the high mortality rate in these mice within the first two weeks makes it challenging to study the role of Runx3 in colitis. To address this issue, a spontaneous colitis (SC) mouse model carrying a C-terminal truncated form of Runx3 with Tyr319stop point mutation has been generated. After weaning, SC mice developed spontaneous diarrhea and exhibited prominent enlargement of the colon, accompanied by severe inflammatory cell infiltration. Results of immunofluorescence staining showed massive CD4+ T cell infiltration in the inflammatory colon of SC mice. Colonic IL-17A mRNA expression and serum IL-17A level were increased in SC mice. CD4+ T cells from SC mice produced stronger IL-17A than those from wildtype mice in Th17-skewing conditions in vitro. In addition, the percentages of Foxp3+ Treg cells as well as the RORγt+Foxp3+ Treg subset, known for its role in suppressing Th17 response in the gut, were notably lower in colon lamina propria of SC mice than those in WT mice. Furthermore, transfer of total CD4+ T cells from SC mice, but not from wildtype mice, into Rag1-ko host mice resulted in severe autoimmune colitis. In conclusion, the C-terminal truncated Runx3 caused autoimmune colitis associated with Th17/Treg imbalance. The SC mouse model is a feasible approach to investigate the effect of immune response on spontaneous colitis.


Asunto(s)
Colitis , Subunidad alfa 3 del Factor de Unión al Sitio Principal , Modelos Animales de Enfermedad , Linfocitos T Reguladores , Células Th17 , Animales , Células Th17/inmunología , Linfocitos T Reguladores/inmunología , Ratones , Colitis/inmunología , Colitis/inducido químicamente , Colitis/genética , Colitis/etiología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Ratones Noqueados , Humanos , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/etiología , Ratones Endogámicos C57BL , Interleucina-17/metabolismo , Interleucina-17/genética , Colon/patología , Colon/inmunología
8.
PLoS One ; 19(4): e0300789, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38625861

RESUMEN

PURPOSE: Immunotherapy has been shown to improve cancer survival, but there are no consensus guidelines to inform use in patients with both cancer and autoimmune disease (AD). We sought to examine immunotherapy utilization patterns between cancer patients with and without AD. PATIENTS AND METHODS: This retrospective cohort study utilized data from a de-identified nationwide oncology database. Patients diagnosed with advanced melanoma, non-small cell lung cancer, and renal cell carcinoma were included. Outcomes of interest included first-line immunotherapy, overall immunotherapy, and number of immunotherapy cycles. We used logistic and Poisson regression models to examine associations between AD and immunotherapy utilization patterns. RESULTS: A total of 25,076 patients were included (796 with AD). Patients with AD were more likely to be female, White, receive care at academic centers, and have ECOG ≥ 3. Controlling for demographic and clinical variables, AD was associated with lower odds of receiving first-line (odds ratio [OR] = 0.68, 95% confidence interval [CI] 0.56-0.82) and overall (OR = 0.80, 95% CI 0.67-0.94) immunotherapy. Among patients who received at least one cycle of immunotherapy, there was no difference in mean number of cycles received between patients with and without AD (11.3 and 10.5 cycles respectively). The incident rate of immunotherapy cycles received for patients with AD was 1.03 times that of patients without AD (95% CI 1.01-1.06). DISCUSSION: Patients with AD were less likely to receive immunotherapy as first-line and overall therapy for treatment of their advanced cancer. However, among those who did receive at least one cycle of immunotherapy, patients with AD received a similar number of cycles compared to patients without AD. This not only indicates that AD is not an absolute contraindication for immunotherapy in clinical practice but may also demonstrate overall treatment tolerability and net benefit in patients with AD.


Asunto(s)
Enfermedades Autoinmunes , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Renales , Neoplasias Pulmonares , Humanos , Femenino , Masculino , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Estudios Retrospectivos , Inmunoterapia/efectos adversos , Neoplasias Renales/etiología , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/etiología
9.
Nature ; 626(8000): 827-835, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355791

RESUMEN

Individuals differ widely in their immune responses, with age, sex and genetic factors having major roles in this inherent variability1-6. However, the variables that drive such differences in cytokine secretion-a crucial component of the host response to immune challenges-remain poorly defined. Here we investigated 136 variables and identified smoking, cytomegalovirus latent infection and body mass index as major contributors to variability in cytokine response, with effects of comparable magnitudes with age, sex and genetics. We find that smoking influences both innate and adaptive immune responses. Notably, its effect on innate responses is quickly lost after smoking cessation and is specifically associated with plasma levels of CEACAM6, whereas its effect on adaptive responses persists long after individuals quit smoking and is associated with epigenetic memory. This is supported by the association of the past smoking effect on cytokine responses with DNA methylation at specific signal trans-activators and regulators of metabolism. Our findings identify three novel variables associated with cytokine secretion variability and reveal roles for smoking in the short- and long-term regulation of immune responses. These results have potential clinical implications for the risk of developing infections, cancers or autoimmune diseases.


Asunto(s)
Inmunidad Adaptativa , Fumar , Femenino , Humanos , Masculino , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/genética , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Índice de Masa Corporal , Citocinas/sangre , Citocinas/inmunología , Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Infecciones/etiología , Infecciones/inmunología , Neoplasias/etiología , Neoplasias/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Fumar/efectos adversos , Fumar/sangre , Fumar/genética , Fumar/inmunología
11.
RMD Open ; 9(4)2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996128

RESUMEN

Autoimmune disorders occur when immune cells go wrong and attack the body's own tissues. Currently, autoimmune disorders are largely treated by broad immunosuppressive agents and blocking antibodies, which can manage the diseases but often are not curative. Thus, there is an urgent need for advanced therapies for patients suffering from severe and refractory autoimmune diseases, and researchers have considered cell therapy as potentially curative approach for several decades. In the wake of its success in cancer therapy, adoptive transfer of engineered T cells modified with chimeric antigen receptors (CAR) for target recognition could now become a therapeutic option for some autoimmune diseases. Here, we review the ongoing developments with CAR T cells in the field of autoimmune disorders. We will cover first clinical results of applying anti-CD19 and anti-B cell maturation antigen CAR T cells for B cell elimination in systemic lupus erythematosus, refractory antisynthetase syndrome and myasthenia gravis, respectively. Furthermore, in preclinical models, researchers have also developed chimeric autoantibody receptor T cells that can eliminate individual B cell clones producing specific autoantibodies, and regulatory CAR T cells that do not eliminate autoreactive immune cells but dampen their wrong activation. Finally, we will address safety and manufacturing aspects for CAR T cells and discuss mRNA technologies and automation concepts for ensuring the future availability of safe and efficient CAR T cell products.


Asunto(s)
Enfermedades Autoinmunes , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Receptores Quiméricos de Antígenos/genética , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/etiología
12.
Int J Mol Sci ; 24(22)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38003230

RESUMEN

Mesenchymal stem cells (MSCs) modulate immune responses and maintain self-tolerance. Their trophic activities and regenerative properties make them potential immunosuppressants for treating autoimmune and autoinflammatory diseases. MSCs are drawn to sites of injury and inflammation where they can both reduce inflammation and contribute to tissue regeneration. An increased understanding of the role of MSCs in the development and progression of autoimmune disorders has revealed that MSCs are passive targets in the inflammatory process, becoming impaired by it and exhibiting loss of immunomodulatory activity. MSCs have been considered as potential novel cell therapies for severe autoimmune and autoinflammatory diseases, which at present have only disease modifying rather than curative treatment options. MSCs are emerging as potential therapies for severe autoimmune and autoinflammatory diseases. Clinical application of MSCs in rare cases of severe disease in which other existing treatment modalities have failed, have demonstrated potential use in treating multiple diseases, including rheumatoid arthritis, systemic lupus erythematosus, myocardial infarction, liver cirrhosis, spinal cord injury, multiple sclerosis, and COVID-19 pneumonia. This review explores the biological mechanisms behind the role of MSCs in autoimmune and autoinflammatory diseases. It also covers their immunomodulatory capabilities, potential therapeutic applications, and the challenges and risks associated with MSC therapy.


Asunto(s)
Enfermedades Autoinmunes , Enfermedades Autoinflamatorias Hereditarias , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/patología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/terapia , Inflamación/terapia , Inflamación/patología , Tolerancia Inmunológica , Inmunomodulación
13.
Mult Scler ; 29(14): 1831-1840, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37830337

RESUMEN

BACKGROUND: Infections are discussed as risk factor for multiple sclerosis (MS) development and relapses. This may lead to decreased vaccination frequency in newly diagnosed patients. OBJECTIVE: The aim of this study was to evaluate the relation of MS diagnosis to subsequent vaccination frequency. METHODS: Based on German ambulatory claims data from 2005 to 2019, regression models were used to assess the relation of MS diagnosis (n = 12,270) to vaccination. A cohort of patients with MS was compared to control cohorts with Crohn's disease, psoriasis, and without these autoimmune diseases (total n = 198,126) in the 5 years after and before diagnosis. RESULTS: Patients with MS were less likely to be vaccinated compared to persons without the autoimmune diseases 5 years after diagnosis (odds ratio = 0.91, p < 0.001). Exceptions were vaccinations against influenza (1.29, p < 0.001) and pneumococci (1.41, p < 0.001). Differences were strong but less pronounced after than before diagnosis (p < 0.001). The likelihood of vaccination was also lower compared to patients with Crohn's disease or psoriasis. CONCLUSIONS: Patients with MS were not adequately vaccinated despite guideline recommendations. Increasing awareness about the importance of vaccination is warranted to reduce the risk of infection, in particular, in patients with MS receiving immunotherapies.


Asunto(s)
Enfermedades Autoinmunes , Enfermedad de Crohn , Esclerosis Múltiple , Psoriasis , Humanos , Enfermedades Autoinmunes/etiología , Factores de Riesgo , Psoriasis/complicaciones , Vacunación/efectos adversos
14.
Bone Marrow Transplant ; 58(11): 1223-1228, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37604871

RESUMEN

Rheumatic autoimmune diseases not only involve the production of autoantibodies but also demonstrate T-cell dysfunction. In patients with concurrent B-cell non-Hodgkin lymphoma (NHL) and rheumatic autoimmune diseases, the safety and efficacy of CD19-targeted chimeric antigen receptor (CAR) T-cell therapy are unknown. Using an aggregated electronic health record database, patients with rheumatic autoimmune diseases (auto group) were compared to propensity score-matched patients without rheumatic autoimmune diseases (non-auto group). From 1/2019 to 1/2023, 58 (4.3%) of 1,363 patients who received CD19-targeted CAR T-cell therapy had concurrent rheumatic autoimmune diseases. Both groups had similar incidence, severity, and management of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Moreover, the two groups had similar time-to-next treatment or death (hazard ratio [HR] 0.97, 95% confidence interval [CI] 0.60 to 1.59, log-rank p = 0.91) and overall survival (HR 0.90, 95%CI 0.46 to 1.78, p = 0.76). Following CAR T-cell infusion, patients with rheumatic autoimmune diseases achieved decreased inflammatory markers, seronegative conversion of autoantibodies, as well as reduced use of steroids and disease-modifying anti-rheumatic drugs. In conclusion, the safety and efficacy of CAR T-cell therapy were not affected in patients with rheumatic autoimmune diseases. Moreover, they achieved better biochemical control of underlying rheumatic diseases.


Asunto(s)
Enfermedades Autoinmunes , Linfoma no Hodgkin , Receptores Quiméricos de Antígenos , Humanos , Inmunoterapia Adoptiva/efectos adversos , Receptores de Antígenos de Linfocitos T , Puntaje de Propensión , Antígenos CD19 , Linfoma no Hodgkin/terapia , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/etiología , Autoanticuerpos , Tratamiento Basado en Trasplante de Células y Tejidos
15.
J Neuroimmunol ; 382: 578156, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37556888

RESUMEN

We reported a 61-year-old man presented with 10-month progressing left sciatic neuropathy and 10-day right facial neuropathy. Serum amphiphysin-IgG was positive. 18F-FDG PET/CT of the whole body showed no signs of malignancy. Treatment with plasma exchange and oral prednisone relieved the symptoms. Nine months later, right hemiparesis and seizure of right limbs developed. 18F-FDG and 18F-PBR06 (18 kDa translocator protein, TSPO) radioligand PET/MRI of the whole body revealed intense uptake in the intracranial lesions. Intracranial lymphoma was diagnosed by stereotactic needle brain biopsy. Mononeuropathies could be paraneoplastic syndromes. TSPO shows high uptake in intracranial lymphoma on 18F-PBR06 PET images.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Enfermedades del Nervio Facial , Linfoma , Neuropatía Ciática , Humanos , Masculino , Persona de Mediana Edad , Encéfalo/inmunología , Enfermedades del Nervio Facial/etiología , Enfermedades del Nervio Facial/inmunología , Enfermedades del Nervio Facial/terapia , Fluorodesoxiglucosa F18 , Inmunoglobulina G/inmunología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Radiofármacos , Receptores de GABA/metabolismo , Neuropatía Ciática/etiología , Neuropatía Ciática/inmunología , Neuropatía Ciática/terapia , Neoplasias del Sistema Nervioso Central/complicaciones , Neoplasias del Sistema Nervioso Central/diagnóstico por imagen , Neoplasias del Sistema Nervioso Central/inmunología , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Linfoma/complicaciones , Linfoma/diagnóstico por imagen , Linfoma/inmunología , Polineuropatía Paraneoplásica/etiología , Polineuropatía Paraneoplásica/inmunología , Prednisona/uso terapéutico , Glucocorticoides/uso terapéutico , Intercambio Plasmático , Proteínas del Tejido Nervioso/inmunología
16.
Hematol Oncol Stem Cell Ther ; 17(1): 29-36, 2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37581466

RESUMEN

BACKGROUND AND OBJECTIVES: A hematopoietic stem cell transplant (HSCT) includes a conditioning regimen which may cause unwanted metabolic changes. We analyzed the changes in electrolytes, glucose, urea, and glomerular filtration rate in patients with multiple sclerosis (MS) who underwent an autologous HSCT employing the "Mexican method." PATIENTS AND METHODS: Serum and urinary electrolytes, blood glucose, creatinine, uric acid, and estimated glomerular filtration rate (eGFR) were prospectively assessed on days -11, -9, and 0 in a group of 75 patients with MS receiving an autologous HSCT employing the "Mexican method," which includes high doses of both cyclophosphamide (Cy, 200 mg/kg) and rituximab (1000 mg). RESULTS: The median age of the patients was 46 years, with a range of 20-65. Baseline data were defined at day -11 of the HSCT. There were significant changes in serum and urinary electrolytes, which diminished substantially after the delivery of high-dose Cy; 12 patients (16%) developed hyponatremia and 2 had hyponatremia-induced seizures, which resulted in hospital admissions. A comparison of baseline blood metabolites with those obtained after the full Cy dosage (day 0) revealed a significant increase in blood glucose and uric acid levels with an associated decrease in serum calcium, sodium, and potassium levels. The salient findings were drug-induced hyponatremia and hyperglycemia. CONCLUSION: Significant changes in serum electrolytes, blood glucose, creatinine, uric acid, and estimated glomerular filtration rate (eGFR) were observed in patients given autologous HSCT for MS employing high-dose Cy. Some of these changes may have clinical consequences, mainly those derived from iatrogenic hyponatremia. No evidence of damage to renal function was observed at day 0.


Asunto(s)
Enfermedades Autoinmunes , Trasplante de Células Madre Hematopoyéticas , Hiponatremia , Adulto , Anciano , Humanos , Persona de Mediana Edad , Adulto Joven , Enfermedades Autoinmunes/etiología , Glucemia , Creatinina , Ciclofosfamida/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Hiponatremia/inducido químicamente , Estudios Prospectivos , Acondicionamiento Pretrasplante/métodos , Trasplante Autólogo , Ácido Úrico
17.
Int J Mol Sci ; 24(12)2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37373021

RESUMEN

After Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) developed into a global pandemic, not only the infection itself but also several immune-mediated side effects led to additional consequences. Immune reactions such as epitope spreading and cross-reactivity may also play a role in the development of long-COVID, although the exact pathomechanisms have not yet been elucidated. Infection with SARS-CoV-2 can not only cause direct damage to the lungs but can also lead to secondary indirect organ damage (e.g., myocardial involvement), which is often associated with high mortality. To investigate whether an immune reaction against the viral peptides can lead to organ affection, a mouse strain known to be susceptible to the development of autoimmune diseases, such as experimental autoimmune myocarditis (EAM), was used. First, the mice were immunized with single or pooled peptide sequences of the virus's spike (SP), membrane (MP), nucleocapsid (NP), and envelope protein (EP), then the heart and other organs such as the liver, kidney, lung, intestine, and muscle were examined for signs of inflammation or other damage. Our results showed no significant inflammation or signs of pathology in any of these organs as a result of the immunization with these different viral protein sequences. In summary, immunization with different SARS-CoV-2 spike-, membrane-, nucleocapsid-, and envelope-protein peptides does not significantly affect the heart or other organ systems adversely, even when using a highly susceptible mouse strain for experimental autoimmune diseases. This suggests that inducing an immune reaction against these peptides of the SARS-CoV-2 virus alone is not sufficient to cause inflammation and/or dysfunction of the myocardium or other studied organs.


Asunto(s)
Enfermedades Autoinmunes , COVID-19 , Miocarditis , Ratones , Humanos , Animales , SARS-CoV-2 , Autoinmunidad , Miocarditis/etiología , Epítopos , Síndrome Post Agudo de COVID-19 , Péptidos , Enfermedades Autoinmunes/etiología , Inflamación
18.
Front Immunol ; 14: 1197364, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37342323

RESUMEN

During the past decade, there has been a revolution in cancer therapeutics by the emergence of antibody-based immunotherapies that modulate immune responses against tumors. These therapies have offered treatment options to patients who are no longer responding to classic anti-cancer therapies. By blocking inhibitory signals mediated by surface receptors that are naturally upregulated during activation of antigen-presenting cells (APC) and T cells, predominantly PD-1 and its ligand PD-L1, as well as CTLA-4, such blocking agents have revolutionized cancer treatment. However, breaking these inhibitory signals cannot be selectively targeted to the tumor microenvironment (TME). Since the physiologic role of these inhibitory receptors, known as immune checkpoints (IC) is to maintain peripheral tolerance by preventing the activation of autoreactive immune cells, IC inhibitors (ICI) induce multiple types of immune-related adverse effects (irAEs). These irAEs, together with the natural properties of ICs as gatekeepers of self-tolerance, have precluded the use of ICI in patients with pre-existing autoimmune diseases (ADs). However, currently accumulating data indicates that ICI might be safely administered to such patients. In this review, we discuss mechanisms of well established and newly recognized irAEs and evolving knowledge from the application of ICI therapies in patients with cancer and pre-existing ADs.


Asunto(s)
Antineoplásicos Inmunológicos , Enfermedades Autoinmunes , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Neoplasias , Humanos , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inmunoterapia/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Enfermedades Autoinmunes/etiología , Microambiente Tumoral
19.
Rev. cuba. oftalmol ; 36(2)jun. 2023.
Artículo en Español | LILACS, CUMED | ID: biblio-1550922

RESUMEN

El síndrome de Vogt-Koyanagi-Harada es una enfermedad autoinmune multisistémica crónica, caracterizada por panuveítis difusa granulomatosa bilateral con desprendimiento exudativo de retina y papilitis. Compromete el sistema nervioso central (meninges, disacusia neurosensorial) así como piel y mucosas. A pesar de ser una enfermedad compleja y poco frecuente, se hace necesario comprender la importancia del diagnóstico rápido y el tratamiento oportuno con seguimiento especializado. Es por ello que se decidió realizar una revisión de la literatura con el objetivo de actualizar los conocimientos existentes sobre este tema. La búsqueda se realizó en diferentes publicaciones y textos básicos de la especialidad. Las fuentes consultadas fueron las bases de datos PubMed y Google Scholar. El diagnóstico de la enfermedad es esencialmente clínico y son los oftalmólogos quienes más lo sospechan por ser los síntomas oculares los más frecuentes y dramáticos. El pronóstico visual de los pacientes es generalmente bueno si el diagnóstico es precoz y se indica un tratamiento adecuado. Los corticosteroides sistémicos a altas dosis asociados a inmunosupresores y agentes biológicos tienen gran impacto en la evolución de la enfermedad, sobre todo estos últimos a nivel mundial, previniendo complicaciones y permitiendo resultados visuales satisfactorios para una mejor calidad de vida del paciente(AU)


Vogt-Koyanagi-Harada syndrome is a chronic multisystem autoimmune disease characterized by bilateral diffuse granulomatous panuveitis with exudative retinal detachment and papillitis. It involves the central nervous system (meninges, sensorineural dysacusis) as well as skin and mucous membranes. In spite of being a complex and infrequent disease, it is necessary to understand the importance of rapid diagnosis and timely treatment with specialized follow-up. For this reason, it was decided to carry out a review of the literature with the aim of updating the existing knowledge on this subject. The search was carried out in different publications and basic texts of the specialty. The sources consulted were the PubMed and Google Scholar databases. The diagnosis of the disease is essentially clinical and it is the ophthalmologists who suspect it the most because the ocular symptoms are the most frequent and dramatic. The visual prognosis of patients is generally good if the diagnosis is early and adequate treatment is indicated. Systemic corticosteroids at high doses associated with immunosuppressants and biological agents have a great impact on the evolution of the disease, especially the latter worldwide, preventing complications and allowing satisfactory visual results for a better quality of life of the patient(AU)


Asunto(s)
Humanos , Enfermedades Autoinmunes/etiología , Desprendimiento de Retina/diagnóstico por imagen , Panuveítis/diagnóstico , Corticoesteroides/uso terapéutico , Literatura de Revisión como Asunto
20.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(3): 444-454, 2023 Mar 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-37164928

RESUMEN

Membrane-bound programmed cell death-1 (mPD-1) and membrane-bound programmed cell death-ligands (mPD-Ls) have soluble forms, which are soluble programmed cell death-1 (sPD-1) and soluble programmed cell death-ligands (sPD-Ls) [including soluble programmed cell death-ligand 1 (sPD-L1) and soluble programmed cell death-ligand 2 (sPD-L2)]. sPD-1 and sPD-L2 are mainly produced by alternative splicing isoforms of PD-1 mRNA, while sPD-L1 is produced by matrix metalloproteinases (MMPs) cutting membrane-bound programmed cell death-ligand 1 (mPD-L1). sPD-1 and sPD-Ls play an important role in autoimmune regulation via blocking the mPD-1 /mPD-L1 pathway, while connective tissue disease (CTD) is a kind of disease caused by autoimmune reaction, and abnormal function of mPD-1/mPD-L1 can occur in the occurrence and development of many autoimmune diseases. Therefore, sPD-1 and sPD-Ls play an important role in the pathogenesis of CTD caused by autoimmune reaction via blocking the mPD-1 /mPD-L1 pathway. It is of great practical significance to understand clinical value of sPD-1 and sPD-Ls in various CTDs for improving the quality of life of patients and the underlying mechanism.


Asunto(s)
Enfermedades Autoinmunes , Enfermedades del Tejido Conjuntivo , Humanos , Ligandos , Calidad de Vida , Enfermedades Autoinmunes/etiología , Apoptosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA