Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.723
Filtrar
1.
Rheumatology (Oxford) ; 63(1): 127-139, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-37084267

RESUMEN

OBJECTIVES: Disease flares in the post-coronavirus disease 2019 (COVID-19) vaccination period represent a prominent concern, though risk factors are poorly understood. We studied these flares among patients with idiopathic inflammatory myopathies (IIMs) and other autoimmune rheumatic diseases (AIRDs). METHODS: The COVAD-1 and -2 global surveys were circulated in early 2021 and 2022, respectively, and we captured demographics, comorbidities, AIRDs details, COVID-19 infection history and vaccination details. Flares of IIMs were defined as (a) patient self-reported, (b) immunosuppression (IS) denoted, (c) clinical sign directed and (d) with >7.9-point minimal clinically significant improvement difference worsening of Patient-Reported Outcomes Measurement Information System (PROMIS) PROMISPF10a score. Risk factors of flares were analysed using regression models. RESULTS: Of 15 165 total respondents, 1278 IIMs (age 63 years, 70.3% female, 80.8% Caucasians) and 3453 AIRDs were included. Flares of IIM were seen in 9.6%, 12.7%, 8.7% and 19.6% patients by definitions (a) to (d), respectively, with a median time to flare of 71.5 (10.7-235) days, similar to AIRDs. Patients with active IIMs pre-vaccination (OR 1.2; 95% CI 1.03, 1.6, P = 0.025) were prone to flares, while those receiving rituximab (OR 0.3; 95% CI 0.1, 0.7, P = 0.010) and AZA (OR 0.3, 95% CI 0.1, 0.8, P = 0.016) were at lower risk. Female gender and comorbidities predisposed to flares requiring changes in IS. Asthma (OR 1.62; 95% CI 1.05, 2.50, P = 0.028) and higher pain visual analogue score (OR 1.19; 95% CI 1.11, 1.27, P < 0.001) were associated with disparity between self-reported and IS-denoted flares. CONCLUSION: A diagnosis of IIMs confers an equal risk of flares in the post-COVID-19 vaccination period to AIRDs, with active disease, female gender and comorbidities conferring a higher risk. Disparity between patient- and physician-reported outcomes represents a future avenue for exploration.


Asunto(s)
Enfermedades Autoinmunes , Vacunas contra la COVID-19 , COVID-19 , Miositis , Enfermedades Reumáticas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Autoinmunes/fisiopatología , COVID-19/prevención & control , Vacunas contra la COVID-19/efectos adversos , Miositis/fisiopatología , Encuestas y Cuestionarios , Vacunación/efectos adversos , Progresión de la Enfermedad , Enfermedades Reumáticas/fisiopatología
3.
Rev Endocr Metab Disord ; 24(2): 191-205, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36509987

RESUMEN

Bone marrow contains resident cellular components that are not only involved in bone maintenance but also regulate hematopoiesis and immune responses. The immune system and bone interact with each other, coined osteoimmunology. Hashimoto's thyroiditis (HT) is one of the most common chronic autoimmune diseases which is accompanied by lymphocytic infiltration. It shows elevating thyroid autoantibody levels at an early stage and progresses to thyroid dysfunction ultimately. Different effects exert on bone metabolism during different phases of HT. In this review, we summarized the mechanisms of the long-term effects of HT on bone and the relationship between thyroid autoimmunity and osteoimmunology. For patients with HT, the bone is affected not only by thyroid function and the value of TSH, but also by the setting of the autoimmune background. The autoimmune background implies a breakdown of the mechanisms that control self-reactive system, featuring abnormal immune activation and presence of autoantibodies. The etiology of thyroid autoimmunity and osteoimmunology is complex and involves a number of immune cells, cytokines and chemokines, which regulate the pathogenesis of HT and osteoporosis at the same time, and have potential to affect each other. In addition, vitamin D works as a potent immunomodulator to influence both thyroid immunity and osteoimmunology. We conclude that HT affects bone metabolism at least through endocrine and immune pathways.


Asunto(s)
Huesos , Enfermedad de Hashimoto , Enfermedad de Hashimoto/inmunología , Enfermedad de Hashimoto/metabolismo , Enfermedad de Hashimoto/fisiopatología , Huesos/inmunología , Huesos/metabolismo , Huesos/fisiopatología , Humanos , Glándula Tiroides/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/fisiopatología , Hormonas Tiroideas/metabolismo , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Vitamina D/inmunología , Vitamina D/metabolismo , Animales , Autoinmunidad , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/fisiopatología
4.
Cell Immunol ; 376: 104532, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35537322

RESUMEN

Autoimmune diseases constitute a heterogeneous group of disorders with one common feature - the loss of immune tolerance towards autoantigens. Due to the complexity of the pathogenesis of these diseases, there are still many open questions regarding their etiology. Therefore, scientists unceasingly search for new data hoping to detect dependable biomarkers and design safe and effective treatment. The research on immune checkpoints is in line with these scientific and clinical demands. Immune checkpoints may be the key to understanding the pathogenesis of many immunological disorders. BTLA-HVEM complex, the inhibitory immune checkpoint, has recently caught scientific attention as an important regulator in different immune contexts, including autoreactivity. So far, the BTLA-HVEM complex has been mainly studied in the context of cancer, but as numerous data show, it may also be a target in the treating of autoimmune diseases. In this review, we intend to focus on the mechanisms of BTLA-HVEM interactions in immune cells and summarize the available data in the context of autoimmunity.


Asunto(s)
Enfermedades Autoinmunes , Receptores Inmunológicos , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Autoinmunidad/inmunología , Humanos , Complejos Multiproteicos/inmunología , Receptores Inmunológicos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología
5.
Front Immunol ; 13: 796054, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154112

RESUMEN

HLA-G is a non-classical HLA class I molecule with immunomodulatory properties. It was initially described at the maternal-fetal interface, and it was later found that this molecule was constitutively expressed on certain immuneprivileged tissues, such as cornea, endothelial and erythroid precursors, and thymus. The immunosuppressive effect of HLA-G is exerted through the interaction with its cognate receptors, expressed on immunocompetent cells, like ILT2, expressed on NK, B, T cells and APCs; ILT4, on APCs; KIR, found on the surface of NK cells; and finally, the co-receptor CD8. Because of these immunomodulatory functions, HLA-G has been involved in several processes, amongst which organ transplantation, viral infections, cancer progression, and autoimmunity. HLA-G neo-expression on tumors has been recently described in several types of malignancies. In fact, tumor progression is tightly linked to the presence of the molecule, as it exerts its tolerogenic function, inhibiting the cells of the immune system and favoring tumor escape. Several polymorphisms in the 3'UTR region condition changes in HLA-G expression (14bp and +3142C/G, among others), which have been associated with both the development and outcome of patients with different tumor types. Also, in recent years, several studies have shown that HLA-G plays an important role in the control of autoimmune diseases. The ability of HLA-G to limit the progression of these diseases has been confirmed and, in fact, levels of the molecule and several of its polymorphisms have been associated with increased susceptibility to the development of autoimmune diseases, as well as increased disease severity. Thus, modulating HLA-G expression in target tissues of oncology patients or patients with autoimmune diseases may be potential therapeutic approaches to treat these pathological conditions.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Antígenos HLA-G/genética , Antígenos HLA-G/inmunología , Neoplasias/inmunología , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/fisiopatología , Humanos , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Ratones , Neoplasias/etiología , Neoplasias/fisiopatología , Polimorfismo Genético , Linfocitos T/inmunología
6.
Dis Markers ; 2022: 4802480, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126787

RESUMEN

OBJECTIVE: To investigate the clinical manifestations, laboratory and imaging examinations, and the treatment outcomes of autoimmune encephalitis (AE). METHODS: The clinical data of 13 patients with autoimmune encephalitis who were hospitalized in the department of neurology, Liaocheng People's Hospital from July 2016 to August 2018 were retrospectively analyzed. RESULTS: The average age of onset of the 13 patients was 45 years, including 6 cases (46%) of anti-NMDAR encephalitis, 3 cases (23%) of anti-GABAB receptor encephalitis, and 4 cases (30%) of anti-LG11 encephalitis, and 4 of them showed abnormal signals of brain MRI (30%). 13 patients (100%) had cognitive impairment and psychiatric symptoms; seizures occurred in 12 patients (92%); lung cancer was found in 1 patient (7%). One case was given up because of the treatment of lung cancer, the other was controlled obviously in epilepsy, and cognitive impairment and abnormal mental behavior were also significantly improved. CONCLUSION: Patients with AE still need to be diagnosed early to avoid missed diagnosis and receive early immunosuppressive therapy, which could effectively reduce mortality and morbidity. A detailed history, clinical manifestations, and positive results for specific NSAbs tests can confirm the diagnosis, and the treatment is mainly done by immunosuppressive therapy.


Asunto(s)
Enfermedades Autoinmunes , Encefalitis , Adolescente , Adulto , Anciano , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Encefalitis/inmunología , Encefalitis/patología , Encefalitis/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
7.
Ann Clin Transl Neurol ; 9(1): 67-78, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35015932

RESUMEN

OBJECTIVES: To describe the clinical presentation, investigations, management, and disease course in pediatric autoimmune limbic encephalitis (LE). METHODS: In this retrospective observational study, from the UK Childhood Neuroinflammatory Disease network, we identified children from six tertiary centers with LE <18 years old between 2008 and 2021. Clinical and paraclinical data were retrieved from medical records. RESULTS: Twenty-five children fulfilling LE criteria were identified, with median age of 11 years (IQR 8, 14) and median follow-up of 24 months (IQR 18, 48). All children presented with seizures; 15/25 (60%) were admitted to intensive care. Neuroimaging demonstrated asymmetric mesial temporal changes in 8/25 (32%), and extra-limbic changes with claustrum involvement in 9/25 (38%). None were positive for LGI1/CASPR2 antibodies (Abs), 2/25 were positive for serum anti-NMDAR Abs, and 2/15 positive for anti-Hu Abs; one died from relapsing neuroblastoma. Two children had serum and CSF anti-GAD antibodies. Initial immune therapy included steroids in 23/25 (92%), intravenous immunoglobulin (IVIg) in 14/25 (56%), and plasma exchange in 7/25 (28%). The commonest second-line treatment was rituximab in 15/25 (60%). Median duration of hospital admission was 21 days (IQR 11, 30). At last follow-up, 13/25 (52%) had refractory seizures and 16/25 (64%) had memory impairment. Six children (24%) had modified Rankin Scale (mRS) scores ≥3. There was no significant difference in mRS, or long-term cognitive and epilepsy outcomes in those who received rituximab versus those who did not. INTERPRETATION: A diagnosis of autoimmune LE was associated with significant morbidity and adverse outcomes in this pediatric cohort.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes , Factores Inmunológicos/administración & dosificación , Encefalitis Límbica , Intercambio Plasmático , Adolescente , Autoanticuerpos/sangre , Autoanticuerpos/líquido cefalorraquídeo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Enfermedades Autoinmunes/terapia , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Unidades de Cuidado Intensivo Pediátrico , Encefalitis Límbica/inmunología , Encefalitis Límbica/patología , Encefalitis Límbica/fisiopatología , Encefalitis Límbica/terapia , Masculino , Evaluación de Resultado en la Atención de Salud , Estudios Retrospectivos , Rituximab/administración & dosificación , Convulsiones
8.
Rev. Hosp. Clin. Univ. Chile ; 33(2): 108-119, 2022. tab
Artículo en Español | LILACS | ID: biblio-1401171

RESUMEN

Vogt-Koyanagi-Harada disease (VKH) is an autoimmune multisystemic syndrome that includes bilateral intraocular inflammation, associated with exudative retinal detachments, and systemic manifestations in the auditory, integumentary, and central nervous systems. The frequency of VKH disease in the world is variable, but in Santiago, Chile, it causes approximately 17% of non-infectious uveitis, an incidence 2 to 3-fold greater than in the USA or European countries. The evidence shows that the pathogenesis of VKH would be caused by cell-mediated autoimmunity directed against melanocytes present in the uveal tissue. CD4+ T lymphocytes (especially hyperactivity of Th17 and Th1 cells), B lymphocytes, cytokines (e.g., TGF-ß, IL-2, IL-6, IL-23 and INF-γ) and chemokines appear to play an important role in the development of VKH. Several lines of evidence support that the pathogenesis of uveitis observed in VKH involves an altered pattern of micro-ribonucleic acids (miRNA) expression, driving the loss of immunological tolerance. In this review, we discuss the evidence related to regulation and altered expression of miRNA associated with Vogt-Koyanagi-Harada and other autoimmune diseases. (AU)


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Síndrome Uveomeningoencefálico/fisiopatología , MicroARNs/genética , Enfermedades Autoinmunes/fisiopatología , Síndrome Uveomeningoencefálico/genética , Síndrome Uveomeningoencefálico/epidemiología
9.
Front Immunol ; 12: 738097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899696

RESUMEN

Background: Sleep disorders (SDs) in autoimmune encephalitis (AE) have received little attention and are poorly understood. We investigated the clinical characteristics, risk factors, and cerebral metabolic mechanism of SD in AE. Methods: Clinical, laboratory, and imaging data were retrospectively reviewed in 121 consecutively patients with definite AE. The risk factors for SD in AE were estimated by logistic regression analysis. Group comparisons based on 18F-fluorodeoxy-glucose positron emission tomography (18F-FDG-PET) data were made between patients with and without SD, to further analyze potential brain metabolic mechanism of SD in AE. Results: A total of 52.9% patients (64/121) with SD were identified. The multivariate logistic model analysis showed that smoking [odds ratio (OR), 6.774 (95% CI, 1.238-37.082); p = 0.027], increased Hamilton Depression scale (HAMD) score [OR, 1.074 (95% CI, 1.002-1.152); p = 0.045], hyperhomocysteinemia [OR, 2.815 (95% CI, 1.057-7.496); p = 0.038], elevated neuron-specific enolase (NSE) level [OR, 1.069 (95% CI, 1.007-1.135); p = 0.03] were independently correlated with higher risk of SD in AE patients. Contrastingly, high MoCA score [OR, 0.821 (95% CI, 0.752-0.896); p < 0.001] was associated with lower risk of SD in AE subjects. Compared to controls, AE patients had less total sleep time, less sleep efficiency, longer sleep latency, more wake, higher percent of stage N1, lower percent of stage N3 and rapid eye movement, and more arousal index in non-rapid eye movement sleep (p < 0.05 for all). Voxel-based group comparison analysis showed that, compared to patients without SD, patients with SD had increased metabolism in the basal ganglia, cerebellum, brainstem, median temporal lobe, thalamus, and hypothalamus [p < 0.05, false discovery rate (FDR) corrected]; decreased metabolism in superior frontal gyrus, medial frontal gyrus, and posterior cingulate cortex (p < 0.001, uncorrected). These results were confirmed by region of interest-based analysis between PET and sleep quality. Conclusion: Smoking, increased HAMD score, hyperhomocysteinemia, and elevated NSE level were correlated with higher risk of SD. High MoCA score was associated with lower risk of SD in AE subjects. Moreover, a widespread metabolic network dysfunction may be involved in the pathological mechanism of SD in AE.


Asunto(s)
Enfermedades Autoinmunes/complicaciones , Encefalitis/complicaciones , Trastornos del Sueño-Vigilia/etiología , Adulto , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/fisiopatología , Encefalitis/metabolismo , Encefalitis/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Estudios Retrospectivos , Factores de Riesgo , Trastornos del Sueño-Vigilia/metabolismo , Trastornos del Sueño-Vigilia/fisiopatología
10.
Front Immunol ; 12: 780689, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899744

RESUMEN

A20, encoded by the TNFAIP3 gene, is a negative regulator of tumor necrosis factor (TNF)-nuclear factor-κB signaling. It was recently demonstrated that A20 haploinsufficiency (HA20), caused by a heterozygous mutation in the TNFAIP3 gene, can present as an early onset autoinflammatory disease resembling Behçet's disease (BD). In addition to autoinflammatory symptoms, HA20 was also reported to be associated with autoimmune diseases and immunodeficiency. Because the phenotypes associated with HA20 are broad, with different severities observed even among individuals in the same family with identical mutations, it has been assumed that the symptoms of HA20 may depend on genetic background and environmental factors. In this review, we summarize the characteristics of patients with HA20 in East Asia and compare these with patients in other regions, mainly the USA and Europe. Patients with HA20 in East Asia developed recurrent fever more frequently than patients in other regions, but were less likely to develop typical BD symptoms such as skin rashes and genital ulcers. In addition, patients with HA20 in East Asia had low rates of complication with autoimmune diseases and low autoantibody detection rates. While anti-TNF-α agents were the primary treatments for severe HA20 in East Asia, anti-interleukin-1 agents and Janus kinase inhibitors were also administered in other regions. Future studies will need to establish methods for analyzing the pathophysiology of HA20 and determining optimal treatment strategies for each patient.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Autoinflamatorias Hereditarias/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Asia Oriental , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Enfermedades Autoinflamatorias Hereditarias/inmunología , Enfermedades Autoinflamatorias Hereditarias/fisiopatología , Humanos
11.
Curr Med Sci ; 41(6): 1172-1177, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34893951

RESUMEN

The composition of the gut microbiota, including Akkermansia muciniphila (A. muciniphila), is altered in many neurological diseases and may be involved in the pathophysiological processes of Parkinson's disease (PD). A. muciniphila, a mucin-degrading bacterium, is a potential next-generation microbe that has anti-inflammatory properties and is responsible for keeping the body healthy. As the role of A. muciniphila in PD has become increasingly apparent, we discuss the potential link between A. muciniphila and various neurological diseases (including PD) in the current review.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal , Enfermedad de Parkinson , Akkermansia , Enfermedades Autoinmunes/fisiopatología , Humanos , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/fisiopatología , Verrucomicrobia
12.
Inflammopharmacology ; 29(6): 1683-1699, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34792672

RESUMEN

The ubiquitin-proteasome pathway is responsible for the turnover of different cellular proteins, such as transport proteins, presentation of antigens to the immune system, control of the cell cycle, and activities that promote cancer. The enzymes which remove ubiquitin, deubiquitylating enzymes (DUBs), play a critical role in central and peripheral immune tolerance to prevent the development of autoimmune diseases and thus present a potential therapeutic target for the treatment of autoimmune diseases. DUBs function by removing ubiquitin(s) from target protein and block ubiquitin chain elongation. The addition and removal of ubiquitin molecules have a significant impact on immune responses. DUBs and E3 ligases both specifically cleave target protein and modulate protein activity and expression. The balance between ubiquitylation and deubiquitylation modulates protein levels and also protein interactions. Dysregulation of the ubiquitin-proteasome pathway results in the development of various autoimmune diseases such as inflammatory bowel diseases (IBD), psoriasis, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). This review summarizes the current understanding of ubiquitination in autoimmune diseases and focuses on various DUBs responsible for the progression of autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Enzimas Desubicuitinizantes/metabolismo , Terapia Molecular Dirigida , Animales , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/terapia , Progresión de la Enfermedad , Humanos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología
13.
Front Immunol ; 12: 753789, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659260

RESUMEN

Innate immunity is regulated by a broad set of evolutionary conserved receptors to finely probe the local environment and maintain host integrity. Besides pathogen recognition through conserved motifs, several of these receptors also sense aberrant or misplaced self-molecules as a sign of perturbed homeostasis. Among them, self-nucleic acid sensing by the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway alerts on the presence of both exogenous and endogenous DNA in the cytoplasm. We review recent literature demonstrating that self-nucleic acid detection through the STING pathway is central to numerous processes, from cell physiology to sterile injury, auto-immunity and cancer. We address the role of STING in autoimmune diseases linked to dysfunctional DNAse or related to mutations in DNA sensing pathways. We expose the role of the cGAS/STING pathway in inflammatory diseases, neurodegenerative conditions and cancer. Connections between STING in various cell processes including autophagy and cell death are developed. Finally, we review proposed mechanisms to explain the sources of cytoplasmic DNA.


Asunto(s)
Enfermedades Autoinmunes/inmunología , ADN/análisis , Inmunidad Innata/fisiología , Inflamación/inmunología , Proteínas de la Membrana/fisiología , Neoplasias/inmunología , Enfermedades Neurodegenerativas/inmunología , Adenosina Trifosfato/metabolismo , Adulto , Enfermedades Autoinmunes/fisiopatología , Autofagia , Citocinas/fisiología , Citoplasma/química , Guanosina Trifosfato/metabolismo , Humanos , Lactante , Inflamación/fisiopatología , Interferón Tipo I/fisiología , Mitocondrias/fisiología , FN-kappa B/metabolismo , Neoplasias/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/fisiología , Transducción de Señal/fisiología
14.
Neurobiol Dis ; 160: 105537, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34695575

RESUMEN

Leucine-Rich Glioma Inactivated protein 1 (LGI1) is a secreted neuronal protein highly expressed in the central nervous system and high amount are found in the hippocampus. An alteration of its function has been described in few families of patients with autosomal dominant temporal lobe epilepsy (ADLTE) or with autoimmune limbic encephalitis (LE), both characterized by epileptic seizures. Studies have shown that LGI1 plays an essential role during development, but also in neuronal excitability through an action on voltage-gated potassium Kv1.1 channels, and in synaptic transmission by regulating the surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-R). Over the last decade, a growing number of studies investigating LGI1 functions have been published. They aimed to improve the understanding of LGI1 function in the regulation of neuronal networks using different animal and cellular models. LGI1 appears to be a major actor of synaptic regulation by modulating trans-synaptically pre- and post-synaptic proteins. In this review, we will focus on LGI1 binding partners, "A Disintegrin And Metalloprotease (ADAM) 22 and 23", the complex they form at the synapse, and will discuss the effects of LGI1 on neuronal excitability and synaptic transmission in physiological and pathological conditions. Finally, we will highlight new insights regarding N-terminal Leucine-Rich Repeat (LRR) domain and C-terminal Epitempin repeat (EPTP) domain and their potentially distinct role in LGI1 function.


Asunto(s)
Enfermedades Autoinmunes/genética , Epilepsia del Lóbulo Temporal/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Encefalitis Límbica/genética , Neuronas/metabolismo , Sinapsis/genética , Transmisión Sináptica/fisiología , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/fisiopatología , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/fisiopatología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Encefalitis Límbica/metabolismo , Encefalitis Límbica/fisiopatología , Sinapsis/metabolismo
15.
Cells ; 10(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34685607

RESUMEN

Within the last two decades, there has been increasing evidence that heat-shock proteins can have a differential influence on the immune system. They can either provoke or ameliorate immune responses. This review focuses on outlining the stimulatory as well as the inhibitory effects of heat-shock proteins 27, 40, 70, 65, 60, and 90 in experimental and clinical autoimmune settings.


Asunto(s)
Autoinmunidad/fisiología , Respuesta al Choque Térmico/fisiología , Animales , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/fisiopatología , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Factores Inmunológicos/metabolismo , Fracciones Subcelulares/metabolismo
16.
Drugs ; 81(15): 1795-1802, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34554438

RESUMEN

Anifrolumab (anifrolumab-fnia; Saphnelo™) is a monoclonal antibody antagonist of the type 1 interferon receptor (IFNAR). It is being developed by AstraZeneca (under license from Medarex, now Bristol-Myers Squibb) for the treatment of autoimmune disorders, including systemic lupus erythematosus (SLE) and lupus nephritis, the underlying pathogenesis of which involves type 1 interferon. In July 2021, intravenous anifrolumab was approved in the USA for the treatment of adult patients with moderate to severe SLE who are receiving standard therapy. Anifrolumab (intravenous or subcutaneous) continues to be assessed in clinical studies in SLE in various countries, and the intravenous formulation is under regulatory review in the EU and Japan. This article summarizes the milestones in the development of anifrolumab leading to this first approval for the treatment of moderate to severe SLE.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Adulto , Anticuerpos Monoclonales Humanizados/farmacología , Enfermedades Autoinmunes/fisiopatología , Aprobación de Drogas , Desarrollo de Medicamentos , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/fisiopatología , Nefritis Lúpica/tratamiento farmacológico , Nefritis Lúpica/fisiopatología
17.
Clin Sci (Lond) ; 135(17): 2165-2196, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34533582

RESUMEN

Hypertension is a major risk factor for cardiovascular disease, chronic kidney disease (CKD), and mortality. Troublingly, hypertension is highly prevalent in patients with autoimmune renal disease and hastens renal functional decline. Although progress has been made over the past two decades in understanding the inflammatory contributions to essential hypertension more broadly, the mechanisms active in autoimmune-mediated renal diseases remain grossly understudied. This Review provides an overview of the pathogenesis of each of the major autoimmune diseases affecting the kidney that are associated with hypertension, and describes the current state of knowledge regarding hypertension in these diseases and their management. Specifically, discussion focuses on Systemic Lupus Erythematosus (SLE) and Lupus Nephritis (LN), Immunoglobulin A (IgA) Nephropathy, Idiopathic Membranous Nephropathy (IMN), Anti-Neutrophil Cytoplasmic Antibody (ANCA)-associated glomerulonephritis, and Thrombotic Thrombocytopenic Purpura (TTP). A summary of disease-specific animal models found to exhibit hypertension is also included to highlight opportunities for much needed further investigation of underlying mechanisms and novel therapeutic approaches.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Presión Sanguínea , Hipertensión/inmunología , Enfermedades Renales/inmunología , Riñón/inmunología , Animales , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/fisiopatología , Modelos Animales de Enfermedad , Humanos , Hipertensión/epidemiología , Hipertensión/fisiopatología , Incidencia , Riñón/fisiopatología , Enfermedades Renales/epidemiología , Enfermedades Renales/fisiopatología , Prevalencia , Medición de Riesgo , Factores de Riesgo
18.
Nat Rev Gastroenterol Hepatol ; 18(12): 885-902, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34580480

RESUMEN

Autoimmune diseases, including inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis, have distinct clinical presentations but share underlying patterns of gut microbiome perturbation and intestinal barrier dysfunction. Their potentially common microbial drivers advocate for treatment strategies aimed at restoring appropriate microbiome function, but individual variation in host factors makes a uniform approach unlikely. In this Perspective, we consolidate knowledge on diet-microbiome interactions in local inflammation, gut microbiota imbalance and host immune dysregulation. By understanding and incorporating the effects of individual dietary components on microbial metabolic output and host physiology, we examine the potential for diet-based therapies for autoimmune disease prevention and treatment. We also discuss tools targeting the gut microbiome, such as faecal microbiota transplantation, probiotics and orthogonal niche engineering, which could be optimized using custom dietary interventions. These approaches highlight paths towards leveraging diet for precise engineering of the gut microbiome at a time of increasing autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/terapia , Dieta/métodos , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/fisiopatología , Terapia Combinada , Trasplante de Microbiota Fecal , Humanos , Prebióticos , Prevención Primaria/métodos , Probióticos/uso terapéutico
19.
Nat Rev Rheumatol ; 17(11): 665-677, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34561652

RESUMEN

Morphological and functional analysis of the microcirculation are objective outcome measures that are recommended for use in the presence of clinical signs of altered peripheral blood flow (such as Raynaud phenomenon), which can occur in systemic sclerosis (SSc) and other autoimmune rheumatic diseases. Several advanced non-invasive tools are available for monitoring the microcirculation, including nailfold videocapillaroscopy, which is the best-studied and most commonly used method for distinguishing and quantifying microvascular morphological alterations in SSc. Nailfold videocapillaroscopy can also be used alongside laser Doppler techniques to assist in the early diagnosis and follow-up of patients with dermatomyositis or mixed connective tissue disease. Power Doppler ultrasonography, which has been used for many years to evaluate the vascularity of synovial tissue in rheumatoid arthritis, is another promising tool for the analysis of skin and nailbed capillary perfusion in other autoimmune rheumatic diseases. Other emerging methods include raster-scanning optoacoustic mesoscopy, which offers non-invasive high-resolution 3D visualization of capillaries and has been tested in psoriatic arthritis and SSc. The principle functions and operative characteristics of several non-invasive tools for analysing microvascular changes are outlined in this Review, and the clinical roles of validated or tested imaging methods are discussed for autoimmune rheumatic diseases.


Asunto(s)
Enfermedades Autoinmunes , Microcirculación , Enfermedades Reumáticas/fisiopatología , Esclerodermia Sistémica/fisiopatología , Enfermedades Vasculares/diagnóstico por imagen , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/fisiopatología , Humanos , Enfermedades Reumáticas/complicaciones , Esclerodermia Sistémica/complicaciones , Ultrasonografía Doppler , Enfermedades Vasculares/etiología , Enfermedades Vasculares/fisiopatología
20.
Sci Rep ; 11(1): 16013, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34362956

RESUMEN

With polygenic risk score (PRS) for autoimmune type 1 diabetes (T1D), this study identified T1D cases with low T1D PRS and searched for susceptibility loci in these cases. Our hypothesis is that genetic effects (likely mediated by relatively rare genetic variants) of non-mainstream (or non-autoimmune) T1D might have been diluted in the previous studies on T1D cases in general. Two cohorts for the PRS modeling and testing respectively were included. The first cohort consisted of 3302 T1D cases and 6181 controls, and the independent second cohort consisted of 3297 T1D cases and 6169 controls. Cases with low T1D PRS were identified using PRSice-2 and compared to controls with low T1D PRS by genome-wide association (GWA) test. Thirteen novel genetic loci with high imputation quality (Quality Score r2 > 0.91) were identified of SNPs/SNVs associated with low PRS T1D at genome-wide significance (P ≤ 5.0 × E-08), in addition to 4 established T1D loci, 3 reported loci by our previous study, as well as 9 potential novel loci represented by rare SNVs, but with relatively low imputation quality (Quality Score r2 < 0.90). For the 13 novel loci, 9 regions have been reported of association with obesity related traits by previous GWA studies. Three loci encoding long intergenic non-protein coding RNAs (lncRNA), and 2 loci involved in N-linked glycosylation are also highlighted in this study.


Asunto(s)
Enfermedades Autoinmunes/fisiopatología , Diabetes Mellitus Tipo 1/genética , Sitios Genéticos , Predisposición Genética a la Enfermedad , Intolerancia a la Glucosa/fisiopatología , Obesidad/fisiopatología , Polimorfismo de Nucleótido Simple , Adolescente , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo , Factores de Riesgo , Estados Unidos/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...