Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 5.764
1.
Cell Commun Signal ; 22(1): 262, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715122

Gene editing of living cells has become a crucial tool in medical research, enabling scientists to address fundamental biological questions and develop novel strategies for disease treatment. This technology has particularly revolutionized adoptive transfer cell therapy products, leading to significant advancements in tumor treatment and offering promising outcomes in managing transplant rejection, autoimmune disorders, and inflammatory diseases. While recent clinical trials have demonstrated the safety of tolerogenic dendritic cell (TolDC) immunotherapy, concerns remain regarding its effectiveness. This review aims to discuss the application of gene editing techniques to enhance the tolerance function of dendritic cells (DCs), with a particular focus on preclinical strategies that are currently being investigated to optimize the tolerogenic phenotype and function of DCs. We explore potential approaches for in vitro generation of TolDCs and provide an overview of emerging strategies for modifying DCs. Additionally, we highlight the primary challenges hindering the clinical adoption of TolDC therapeutics and propose future research directions in this field.


Autoimmune Diseases , Dendritic Cells , Humans , Dendritic Cells/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Animals , Gene Editing/methods , Immunotherapy/methods
2.
Front Immunol ; 15: 1349138, 2024.
Article En | MEDLINE | ID: mdl-38720903

Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.


Autoimmune Diseases , Induced Pluripotent Stem Cells , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Drug Discovery , Cell- and Tissue-Based Therapy/methods
3.
Int J Mol Sci ; 25(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38732038

The gut microbiota and short chain fatty acids (SCFA) have been associated with immune regulation and autoimmune diseases. Autoimmune kidney diseases arise from a loss of tolerance to antigens, often with unclear triggers. In this review, we explore the role of the gut microbiome and how disease, diet, and therapy can alter the gut microbiota consortium. Perturbations in the gut microbiota may systemically induce the translocation of microbiota-derived inflammatory molecules such as liposaccharide (LPS) and other toxins by penetrating the gut epithelial barrier. Once in the blood stream, these pro-inflammatory mediators activate immune cells, which release pro-inflammatory molecules, many of which are antigens in autoimmune diseases. The ratio of gut bacteria Bacteroidetes/Firmicutes is associated with worse outcomes in multiple autoimmune kidney diseases including lupus nephritis, MPO-ANCA vasculitis, and Goodpasture's syndrome. Therapies that enhance SCFA-producing bacteria in the gut have powerful therapeutic potential. Dietary fiber is fermented by gut bacteria which in turn release SCFAs that protect the gut barrier, as well as modulating immune responses towards a tolerogenic anti-inflammatory state. Herein, we describe where the current field of research is and the strategies to harness the gut microbiome as potential therapy.


Autoimmune Diseases , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/immunology , Autoimmune Diseases/microbiology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Fatty Acids, Volatile/metabolism , Kidney Diseases/microbiology , Kidney Diseases/immunology , Kidney Diseases/therapy
4.
Autoimmunity ; 57(1): 2351872, 2024 Dec.
Article En | MEDLINE | ID: mdl-38739691

Autophagy is a highly conserved biological process in eukaryotes, which degrades cellular misfolded proteins, damaged organelles and invasive pathogens in the lysosome-dependent manner. Autoimmune diseases caused by genetic elements, environments and aberrant immune responses severely impact patients' living quality and even threaten life. Recently, numerous studies have reported autophagy can regulate immune responses, and play an important role in autoimmune diseases. In this review, we summarised the features of autophagy and autophagy-related genes, enumerated some autophagy-related genes involved in autoimmune diseases, and further overviewed how to treat autoimmune diseases through targeting autophagy. Finally, we outlooked the prospect of relieving and curing autoimmune diseases by targeting autophagy pathway.


Autoimmune Diseases , Autophagy , Humans , Autophagy/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/therapy , Animals , Signal Transduction/immunology , Molecular Targeted Therapy
5.
Brain Nerve ; 76(5): 534-539, 2024 May.
Article Ja | MEDLINE | ID: mdl-38741493

Autoimmune nodopathy (AN), a newly established category of autoimmune disease, refers to an immune-mediated neuropathy associated with development of autoantibodies against membrane proteins, including neurofascin 186, neurofascin 155, contactin-1, and contactin-associated protein 1 located in the nodes of Ranvier or paranodes. Subclass analysis of these autoantibodies reveals predominant elevation of immunoglobulin (G4. Patients with AN show clinical and laboratory characteristics such as distal-predominant sensorimotor disturbance, sensory ataxia, poor response to intravenous immunoglobulin, and highly elevated cerebrospinal fluid protein levels. B cell-depletion therapy using an anti-CD20 monoclonal antibody is effective for patients with AN. Autoantibody measurement is beneficial not only for diagnosis but also for deciding treatment strategies for AN.


Autoantibodies , Humans , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/therapy
6.
Front Immunol ; 15: 1385190, 2024.
Article En | MEDLINE | ID: mdl-38711523

The discovery of Suppressor of Cytokine Signaling 1 (SOCS1) in 1997 marked a significant milestone in understanding the regulation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathways. Subsequent research deciphered its cellular functions, and recent insights into SOCS1 deficiencies in humans underscored its critical role in immune regulation. In humans, SOCS-haploinsufficiency (SOCS1-HI) presents a diverse clinical spectrum, encompassing autoimmune diseases, infection susceptibility, and cancer. Variability in disease manifestation, even within families sharing the same genetic variant, raises questions about clinical penetrance and the need for individualized treatments. Current therapeutic strategies include JAK inhibition, with promising results in controlling inflammation in SOCS1-HI patients. Hematopoietic stem cell transplantation and gene therapy emerge as promising avenues for curative treatments. The evolving landscape of SOCS1 research, emphasizes the need for a nuanced understanding of genetic variants and their functional consequences.


Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Humans , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Janus Kinases/metabolism , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Haploinsufficiency , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Genetic Therapy
9.
Front Immunol ; 15: 1366101, 2024.
Article En | MEDLINE | ID: mdl-38707905

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.


Autoimmune Diseases , Glucocorticoids , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Middle Aged , Glucocorticoids/therapeutic use , Autoimmune Diseases/etiology , Autoimmune Diseases/therapy , Rituximab/therapeutic use , Anemia, Hemolytic, Autoimmune/etiology , Anemia, Hemolytic, Autoimmune/therapy , Anemia, Hemolytic, Autoimmune/drug therapy , Drug Resistance
10.
Int J Nanomedicine ; 19: 3943-3956, 2024.
Article En | MEDLINE | ID: mdl-38708179

Autoimmune diseases refer to a group of conditions where the immune system produces an immune response against self-antigens, resulting in tissue damage. These diseases have profound impacts on the health of patients. In recent years, with the rapid development in the field of biomedicine, engineered exosomes have emerged as a noteworthy class of biogenic nanoparticles. By precisely manipulating the cargo and surface markers of exosomes, engineered exosomes have gained enhanced anti-inflammatory, immunomodulatory, and tissue reparative abilities, providing new prospects for the treatment of autoimmune diseases. Engineered exosomes not only facilitate the efficient delivery of bioactive molecules including nucleic acids, proteins, and cytokines, but also possess the capability to modulate immune cell functions, suppress inflammation, and restore immune homeostasis. This review mainly focuses on the applications of engineered exosomes in several typical autoimmune diseases. Additionally, this article comprehensively summarizes the current approaches for modification and engineering of exosomes and outlines their prospects in clinical applications. In conclusion, engineered exosomes, as an innovative therapeutic approach, hold promise for the management of autoimmune diseases. However, while significant progress has been made, further rigorous research is still needed to address the challenges that engineered exosomes may encounter in the therapeutic intervention process, in order to facilitate their successful translation into clinical practice and ultimately benefit a broader population of patients.


Autoimmune Diseases , Exosomes , Exosomes/immunology , Humans , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Animals , Nanoparticles/chemistry
11.
Mol Biol Rep ; 51(1): 629, 2024 May 08.
Article En | MEDLINE | ID: mdl-38717637

It has been rediscovered in the last fifteen years that B-cells play an active role in autoimmune etiology rather than just being spectators. The clinical success of B-cell depletion therapies (BCDTs) has contributed to this. BCDTs, including those that target CD20, CD19, and BAFF, were first developed to eradicate malignant B-cells. These days, they treat autoimmune conditions like multiple sclerosis and systemic lupus erythematosus. Particular surprises have resulted from the use of BCDTs in autoimmune diseases. For example, even in cases where BCDT is used to treat the condition, its effects on antibody-secreting plasma cells and antibody levels are restricted, even though these cells are regarded to play a detrimental pathogenic role in autoimmune diseases. In this Review, we provide an update on our knowledge of the biology of B-cells, examine the outcomes of clinical studies employing BCDT for autoimmune reasons, talk about potential explanations for the drug's mode of action, and make predictions about future approaches to targeting B-cells other than depletion.


Autoimmune Diseases , B-Lymphocytes , Lymphocyte Depletion , Humans , B-Lymphocytes/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Lymphocyte Depletion/methods , Antigens, CD20/immunology , Antigens, CD19/immunology , Animals , B-Cell Activating Factor/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy
12.
Front Immunol ; 15: 1346671, 2024.
Article En | MEDLINE | ID: mdl-38698867

IgG4 subclass antibodies represent the rarest subclass of IgG antibodies, comprising only 3-5% of antibodies circulating in the bloodstream. These antibodies possess unique structural features, notably their ability to undergo a process known as fragment-antigen binding (Fab)-arm exchange, wherein they exchange half-molecules with other IgG4 antibodies. Functionally, IgG4 antibodies primarily block and exert immunomodulatory effects, particularly in the context of IgE isotype-mediated hypersensitivity reactions. In the context of disease, IgG4 antibodies are prominently observed in various autoimmune diseases combined under the term IgG4 autoimmune diseases (IgG4-AID). These diseases include myasthenia gravis (MG) with autoantibodies against muscle-specific tyrosine kinase (MuSK), nodo-paranodopathies with autoantibodies against paranodal and nodal proteins, pemphigus vulgaris and foliaceus with antibodies against desmoglein and encephalitis with antibodies against LGI1/CASPR2. Additionally, IgG4 antibodies are a prominent feature in the rare entity of IgG4 related disease (IgG4-RD). Intriguingly, both IgG4-AID and IgG4-RD demonstrate a remarkable responsiveness to anti-CD20-mediated B cell depletion therapy (BCDT), suggesting shared underlying immunopathologies. This review aims to provide a comprehensive exploration of B cells, antibody subclasses, and their general properties before examining the distinctive characteristics of IgG4 subclass antibodies in the context of health, IgG4-AID and IgG4-RD. Furthermore, we will examine potential therapeutic strategies for these conditions, with a special focus on leveraging insights gained from anti-CD20-mediated BCDT. Through this analysis, we aim to enhance our understanding of the pathogenesis of IgG4-mediated diseases and identify promising possibilities for targeted therapeutic intervention.


Autoantibodies , Autoimmune Diseases , Autoimmunity , Immunoglobulin G , Humans , Immunoglobulin G/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Immunoglobulin G4-Related Disease/immunology , Immunoglobulin G4-Related Disease/therapy
13.
Life Sci ; 348: 122686, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38710282

Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.


Autoimmune Diseases , Autoimmunity , Dendritic Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms , Dendritic Cells/immunology , Dendritic Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Inflammasomes/immunology , Inflammasomes/metabolism , Animals , Autoimmunity/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/metabolism , Communicable Diseases/immunology , Communicable Diseases/metabolism , Communicable Diseases/therapy
16.
Int J Rheum Dis ; 27(5): e15182, 2024 May.
Article En | MEDLINE | ID: mdl-38742463

Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.


Autoimmune Diseases , Hematologic Neoplasms , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Rheumatic Diseases , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Rheumatic Diseases/immunology , Rheumatic Diseases/therapy , Receptors, Chimeric Antigen/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Treatment Outcome , T-Lymphocytes/immunology , Animals
17.
Rev Med Suisse ; 20(868): 688-693, 2024 Apr 03.
Article Fr | MEDLINE | ID: mdl-38568061

Cellular therapy using genetically modified T lymphocytes expressing synthetic receptors, known as CAR (Chimeric Antigen Receptor), has revolutionized the treatment of certain hematologic malignancies. This success has led to exploring the same approach in the treatment of severe autoimmune diseases refractory to conventional therapies. Initial results in systemic lupus erythematosus have shown complete remissions that appear to persist over time. Consequently, there is a growing number of ongoing clinical trials. In this review, we discuss the rationale behind the use of CAR-T therapies, the targeted autoimmune diseases, and the associated risks.


La thérapie cellulaire à base de lymphocytes T génétiquement modifiés exprimant des récepteurs synthétiques ou CAR (récepteur antigénique chimérique) a révolutionné le traitement de certaines maladies hémato-oncologiques. Ce succès a conduit à l'exploration de la même approche dans le traitement de maladies auto-immunes sévères et réfractaires aux thérapies conventionnelles. Les premiers résultats obtenus dans le lupus érythémateux systémique ont montré des rémissions complètes semblant persister dans le temps. Nous assistons donc actuellement à une prolifération importante d'essais cliniques. Dans cet article, nous abordons le rationnel derrière l'utilisation des thérapies CAR-T, les maladies auto-immunes ciblées, mais aussi les risques associés.


Autoimmune Diseases , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive , Autoimmune Diseases/therapy , Cell- and Tissue-Based Therapy , Pathologic Complete Response
18.
J Exp Med ; 221(5)2024 May 06.
Article En | MEDLINE | ID: mdl-38587494

Jennifer S. Michaelson, Chief Scientific Officer at Cullinan Oncology, and Patrick A. Baeuerle, scientific advisor to Cullinan Oncology and honorary professor in immunology at Ludwig Maximilians University Munich, discuss the use of CD19-specific T cell-engaging antibody therapies (TCEs) as therapeutics for autoimmune diseases.


Autoimmune Diseases , Humans , Autoimmune Diseases/therapy , Adaptor Proteins, Signal Transducing , Antigens, CD19 , T-Lymphocytes
19.
Rev Med Suisse ; 20(867): 638-647, 2024 Mar 27.
Article Fr | MEDLINE | ID: mdl-38563538

Auto-immune bullous diseases (AIBD) are rare in children. Although their pathogenesis is similar to their adult counterpart, there are differences in the clinical presentation. Moreover certain AIBD prevail at certain ages. There are no guidelines for the treatment of AIBD specific for children. In this review the recent literature is summarised with attention to recent data including diagnostic criteria. We also propose a treatment algorithm.


Les maladies bulleuses auto-immunes (MBAI) sont rares chez les enfants. Bien que la pathogenèse soit similaire à celle de l'adulte, il existe des différences concernant la présentation clinique et la prévalence des MBAI selon l'âge. À ce jour, il n'y a pas de recommandations spécifiques pour leur prise en charge chez l'enfant. Dans cet article, nous présentons une revue des données actuelles, des critères diagnostiques et proposons un algorithme de prise en charge.


Autoimmune Diseases , Skin Diseases, Vesiculobullous , Child , Humans , Algorithms , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Skin Diseases, Vesiculobullous/diagnosis , Skin Diseases, Vesiculobullous/pathology
20.
Curr Neurol Neurosci Rep ; 24(5): 141-150, 2024 May.
Article En | MEDLINE | ID: mdl-38589696

PURPOSE OF REVIEW: Immune-mediated necrotizing myopathy (IMNM), characterized by acute or subacute onset, severe weakness, and elevated creatine kinase levels, poses diagnostic and therapeutic challenges. This article provides a succinct overview of IMNM, including clinical features, diagnostic strategies, and treatment approaches. RECENT FINDINGS: Recent insights highlight the different clinical presentations and therapeutic options of IMNM stratified by autoantibody positivity and type. Additionally, recent findings call into question the reported link between statin use and IMNM. This review synthesizes current knowledge on IMNM, emphasizing its distinct clinical features and challenging management. The evolving understanding of IMNM underscores the need for a comprehensive diagnostic approach that utilizes a growing range of modalities. Early and aggressive immunomodulatory therapy remains pivotal. Ongoing research aims to refine diagnostic tools and therapeutic interventions for this challenging muscle disorder, underscoring the importance of advancing our understanding to enhance patient outcomes.


Autoimmune Diseases , Muscular Diseases , Myositis , Humans , Muscle, Skeletal , Necrosis/diagnosis , Myositis/therapy , Myositis/drug therapy , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Muscular Diseases/diagnosis , Muscular Diseases/therapy , Autoantibodies
...