Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.740
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999928

ABSTRACT

Autoimmune atrophic gastritis is an immune-mediated disease resulting in autoimmune destruction of the specialized acid-producing gastric parietal cells. As a consequence, in autoimmune atrophic gastritis, gastric acid secretion is irreversibly impaired, and the resulting hypochlorhydria leads to the main clinical manifestations and is linked, directly or indirectly, to the long-term neoplastic complications of this disease. In the last few years, autoimmune atrophic gastritis has gained growing interest leading to the acquisition of new knowledge on different aspects of this disorder. Although reliable serological biomarkers are available and gastrointestinal endoscopy techniques have substantially evolved, the diagnosis of autoimmune atrophic gastritis is still affected by a considerable delay and relies on histopathological assessment of gastric biopsies. One of the reasons for the diagnostic delay is that the clinical presentations of autoimmune atrophic gastritis giving rise to clinical suspicion are very different, ranging from hematological to neurological-psychiatric up to gastrointestinal and less commonly to gynecological-obstetric symptoms or signs. Therefore, patients with autoimmune atrophic gastritis often seek advice from physicians of other medical specialties than gastroenterologists, thus underlining the need for increased awareness of this disease in a broad medical and scientific community.


Subject(s)
Achlorhydria , Autoimmune Diseases , Gastritis, Atrophic , Humans , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Gastritis, Atrophic/pathology , Achlorhydria/metabolism , Biomarkers
2.
Front Immunol ; 15: 1416292, 2024.
Article in English | MEDLINE | ID: mdl-38953024

ABSTRACT

The predominant characteristic of autoimmune gastritis (AIG) is corpus-dominant advanced atrophy, which is mostly observed in the middle to late stages. More reports are needed on the endoscopic features of the early stage. In this report, we present two cases of early-stage AIG in which endoscopic examinations showed no atrophy of the gastric mucosa but displayed a transition of collecting venules from a regular to an irregular arrangement. In addition, yellowish-white cobblestone-like elevations were observed in the fundic gland region. Histologically, the observed manifestations included pseudohypertrophy and protrusion of parietal cells into the lumen, possibly along with hyperplasia of G cells, lymphocytic infiltration and potentially pseudopyloric gland metaplasia. Serologically, the anti-parietal cell antibody returned positive results, whereas the anti-intrinsic factor antibody yielded negative results. In this study, we summarized some endoscopic features of two patients, aiming to provide clues for endoscopists to detect early-stage AIG.


Subject(s)
Autoimmune Diseases , Gastritis , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/pathology , Male , Gastritis/immunology , Gastritis/diagnosis , Gastritis/pathology , Female , Middle Aged , Autoantibodies/immunology , Gastric Mucosa/pathology , Gastric Mucosa/immunology , Parietal Cells, Gastric/immunology , Parietal Cells, Gastric/pathology , Gastroscopy , Biopsy , Aged , Adult
3.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891767

ABSTRACT

Myocarditis is characterized by an influx of inflammatory cells, predominantly of myeloid lineage. The progression of myocarditis to a dilated cardiomyopathy is markedly influenced by TGF-ß signalling. Here, we investigate the role of TGF-ß signalling in inflammatory cardiac macrophages in the development of myocarditis and post-inflammatory fibrosis. Experimental autoimmune myocarditis (EAM) was induced in the LysM-Cre × R26-stop-EYFP × Tgfbr2-fl/fl transgenic mice showing impaired TGF-ß signalling in the myeloid lineage and the LysM-Cre × R26-stop-EYFP control mice. In EAM, immunization led to acute myocarditis on day 21, followed by cardiac fibrosis on day 40. Both strains showed a similar severity of myocarditis and the extent of cardiac fibrosis. On day 21 of EAM, an increase in cardiac inflammatory macrophages was observed in both strains. These cells were sorted and analysed for differential gene expression using whole-genome transcriptomics. The analysis revealed activation and regulation of the inflammatory response, particularly the production of both pro-inflammatory and anti-inflammatory cytokines and cytokine receptors as TGF-ß-dependent processes. The analysis of selected cytokines produced by bone marrow-derived macrophages confirmed their suppressed secretion. In conclusion, our findings highlight the regulatory role of TGF-ß signalling in cytokine production within inflammatory cardiac macrophages during myocarditis.


Subject(s)
Autoimmune Diseases , Cytokines , Macrophages , Mice, Transgenic , Myocarditis , Signal Transduction , Transforming Growth Factor beta , Animals , Myocarditis/metabolism , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/etiology , Transforming Growth Factor beta/metabolism , Mice , Macrophages/metabolism , Macrophages/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cytokines/metabolism , Disease Models, Animal , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology , Fibrosis , Male
4.
Head Neck Pathol ; 18(1): 49, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884863

ABSTRACT

BACKGROUND: Oral lesions of immune-mediated and autoimmune diseases have been well-documented, but studies from Brazil are limited. The varied spectrum of oral lesions within this demographic group poses challenges to clinicians, particularly when they occur in isolation. This study aimed to evaluate the occurrence, clinical characteristics, and management of patients with oral lesions of immune-mediated and autoimmune diseases at a single center in Brazil. METHODS: A retrospective cross-sectional study was conducted from 2010 to 2022. Clinicodemographic data, histopathological features, and treatment modalities were analyzed descriptively and analytically. RESULTS: Of the 3,790 oral and maxillofacial lesions diagnosed, 160 (4.2%) were confirmed as immune-mediated or autoimmune diseases. The population surveyed predominantly consisted of women (73.7%), with a mean age of 60.2 years. Oral lichen planus (51.3%), mucous membrane pemphigoid (MMP) (23.7%), and pemphigus vulgaris (PV) (19.4%) were the most prevalent lesions. The buccal mucosa (59.4%) was predominantly affected, with pain reported in 46.2% of cases, notably in individuals with PV and MMP. The average time to disease stabilization post-local and/or systemic corticosteroid therapy was 15.8 months for systemic lupus erythematosus, 8.7 months for MMP, and 6.5 months for PV. CONCLUSION: Although oral lesions related to immune-mediated and autoimmune diseases are uncommon, their diverse clinicopathological aspects require multidisciplinary management.


Subject(s)
Autoimmune Diseases , Mouth Diseases , Humans , Female , Retrospective Studies , Middle Aged , Male , Cross-Sectional Studies , Autoimmune Diseases/pathology , Brazil/epidemiology , Mouth Diseases/pathology , Aged , Adult , Young Adult , Aged, 80 and over , Adolescent
5.
J Clin Exp Hematop ; 64(2): 97-106, 2024.
Article in English | MEDLINE | ID: mdl-38925977

ABSTRACT

Atypical lymphoplasmacytic and immunoblastic proliferation (ALPIBP) was first reported in 1984 as characteristic histological findings in lymph nodes associated with autoimmune diseases, but it has not been clearly defined to date. To summarize the histological characteristics and clinical diagnoses associated with ALPIBP, we searched MEDLINE and EMBASE for all peer-reviewed articles using keywords including "atypical lymphoplasmacytic and immunoblastic lymphadenopathy" from their inception to December 27, 2023. We also summarized the courses of three cases with a pathological diagnosis of ALPIBP. Nine articles with 52 cases were included. Among the total of 55 cases, including the three from our institution, the median age of the cases was 63.5 years with a female predominance (69.5%). Lymphadenopathy was generalized in 65.6% and regional in 34.4% of cases. RA (24.4%), SLE (24.4%), and autoimmune hemolytic anemia (20.0%), were common clinical diagnoses. A combination of cytotoxic chemotherapy was used in 15.6% of cases due to the suspicion of malignancy. Nodal T-follicular helper cell lymphoma, angioimmunoblastic type, methotrexate-associated lymphoproliferative disorders, and IgG4-related diseases were listed as important diseases that need to be pathologically differentiated from ALPIBP. This review summarizes the current understanding of the characteristics of ALPIBP. Given that underrecognition of ALPIBP could lead to overdiagnosis of hematological malignancy and unnecessary treatment, increased awareness of the condition in pathologists and clinicians is crucial.


Subject(s)
Lymphoproliferative Disorders , Humans , Female , Male , Middle Aged , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/diagnosis , Lymphadenopathy/pathology , Lymphadenopathy/diagnosis , Lymph Nodes/pathology , Autoimmune Diseases/pathology , Autoimmune Diseases/diagnosis
6.
Immunopharmacol Immunotoxicol ; 46(4): 529-537, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38918174

ABSTRACT

OBJECTIVE: Autoimmune dermatosis (AID) occurs when the body's immune system attacks skin or tissue, leading to various types of skin disorders or injuries. Recent studies show that Janus kinases (JAKs) play critical roles in autoimmune diseases including AID by regulating multiple cytokine signaling pathways. CS12192, a novel JAK3/JAK1/TBK1 inhibitor, has been reported to exert ameliorative effects in rheumatoid arthritis. However, the efficacy of CS12192 on AID is undetermined. This study aims to investigate the therapeutic efficacy of CS12192 on psoriasis (PSO), systemic lupus erythematosus (SLE) and atopic dermatitis (AD) in mouse models. METHODS: Interleukin-23 (IL-23)-induced PSO model, spontaneous SLE model of MRL/MpJ-Faslpr/J (MRL/lpr) mice, and oxazolone (OXA) and dinitrochlorobenzene (DNCB)-induced murine AD models were used for the evaluation of curative effects of CS12192, respectively. The skin lesion, biochemical parameters, ear thickness, ear weight and histopathology were assessed accordingly. RESULTS: In PSO model, mice treated with CS12192 show reduced ear thickness and ear weight as compared with vehicle. In SLE model, CS12192 ameliorates cutaneous parameters such as lymphadenectasis and skin lesion but not systematic parameters such as proteinuria concentration and score, serum dsDNA and BUN concentration. In AD models, CS12192 dose-dependently improves ear swelling and reduces histological scores, exerting equivalent efficacy with baricitinib, a marketed JAK1/JAK2 inhibitor. CONCLUSION: Our findings suggest that the novel JAK3/JAK1/TBK1 inhibitor CS12192 is potentially to alleviate autoimmune dermatosis.


Subject(s)
Disease Models, Animal , Janus Kinase 1 , Janus Kinase 3 , Protein Serine-Threonine Kinases , Sulfonamides , Animals , Mice , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 3/antagonists & inhibitors , Sulfonamides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Female , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Purines/pharmacology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Azetidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/pathology , Psoriasis/chemically induced , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/pathology , Pyrazoles
7.
Am J Physiol Regul Integr Comp Physiol ; 327(2): R250-R260, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38842512

ABSTRACT

Although the cause of interstitial cystitis/painful bladder syndrome (IC/PBS) remains unknown, autoimmune involvement has been strongly suggested to be a contributing factor. To elucidate the pathophysiology of IC/PBS, we characterized the experimental autoimmune cystitis (EAC) in rats. Adult female Sprague-Dawley rats were divided into the EAC and control groups. The EAC rats were generated by administrating a homogenate of donor rat bladder tissue as a bladder antigen. The characteristics of the two groups were determined by evaluating pain behavior and conducting cystometry, histopathology, and molecular analyses. The EAC rats showed: 1) a decreased paw withdrawal threshold, 2) a reduced intercontraction interval on cystometry, 3) the irregular surfaces of the umbrella cells of epithelium throughout the bladder wall, 4) accumulation of stress granules in the bladder and vascular endothelium, 5)the increased expression of genes related to inflammation and ischemia at the mRNA and protein levels, 6) a significantly increased paw withdrawal threshold with pain treatment, and 7) the induction of glomerulation of the bladder wall, epithelium denudation, and lymphocyte infiltration in the interstitium by bladder distension. These results suggest that the EAC rats showed pain and frequent urination with the overexpression of inflammatory chemokines, reflecting clinical IC/BPS, and the bladder epithelium and vascular endothelium may be the primary sites of IC/BPS, and bladder injury, such as bladder distension, can cause progression from BPS to IC with Hunner lesions.NEW & NOTEWORTHY The experimental autoimmune cystitis model rats showed pain and frequent urination with the overexpression of inflammatory chemokines, reflecting clinical interstitial cystitis/painful bladder syndrome (IC/PBS), and the bladder epithelium and vascular endothelium may be the primary sites of IC/BPS, and bladder injury, such as bladder distension, can cause progression from BPS to IC with Hunner lesions.


Subject(s)
Cystitis, Interstitial , Disease Models, Animal , Rats, Sprague-Dawley , Urinary Bladder , Animals , Cystitis, Interstitial/physiopathology , Cystitis, Interstitial/pathology , Cystitis, Interstitial/metabolism , Cystitis, Interstitial/immunology , Female , Urinary Bladder/physiopathology , Urinary Bladder/pathology , Autoimmune Diseases/physiopathology , Autoimmune Diseases/pathology , Rats , Pain Threshold
8.
Stem Cell Res Ther ; 15(1): 149, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783393

ABSTRACT

BACKGROUND: Autoimmune uveitis is an inflammatory disease triggered by an aberrant immune response. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) are emerging as potential therapeutic agents for this condition. CD73, an ectoenzyme present on MSC-sEVs, is involved in mitigating inflammation by converting extracellular adenosine monophosphate into adenosine. We hypothesize that the inhibitory effect of MSC-sEVs on experimental autoimmune uveitis (EAU) could be partially attributed to the surface expression of CD73. METHODS: To investigate novel therapeutic approaches for autoimmune uveitis, we performed lentiviral transduction to overexpress CD73 on the surface of MSC-sEVs, yielding CD73-enriched MSC-sEVs (sEVs-CD73). Mice with interphotoreceptor retinoid-binding protein (IRBP)-induced EAU were grouped randomly and treated with 50 µg MSC-sEVs, vector infected MSC-sEVs, sEVs-CD73 or PBS via single tail vein injection. We evaluated the clinical and histological features of the induced mice and analyzed the proportion and functional capabilities of T helper cells. Furthermore, T-cells were co-cultured with various MSC-sEVs in vitro, and we quantified the resulting inflammatory response to assess the potential therapeutic benefits of sEVs-CD73. RESULTS: Compared to MSC-sEVs, sEVs-CD73 significantly alleviates EAU, leading to reduced inflammation and diminished tissue damage. Treatment with sEVs-CD73 results in a decreased proportion of Th1 cells in the spleen, draining lymph nodes, and eyes, accompanied by an increased proportion of regulatory T-cells (Treg cells). In vitro assays further reveal that sEVs-CD73 inhibits T-cell proliferation, suppresses Th1 cells differentiation, and enhances Treg cells proportion. CONCLUSION: Over-expression of CD73 on MSC-sEVs enhances their immunosuppressive effects in EAU, indicating that sEVs-CD73 has the potential as an efficient immunotherapeutic agent for autoimmune uveitis.


Subject(s)
5'-Nucleotidase , Autoimmune Diseases , Extracellular Vesicles , Mesenchymal Stem Cells , Uveitis , Animals , Uveitis/pathology , Uveitis/therapy , Uveitis/metabolism , Uveitis/immunology , 5'-Nucleotidase/metabolism , 5'-Nucleotidase/genetics , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Mice , Autoimmune Diseases/therapy , Autoimmune Diseases/pathology , Autoimmune Diseases/immunology , Mice, Inbred C57BL , Disease Models, Animal , Female , Retinol-Binding Proteins , Humans
9.
Kaohsiung J Med Sci ; 40(6): 520-529, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38712483

ABSTRACT

Autoimmune disease is characterized by the proliferation of harmful immune cells, inducing tissue inflammation and ultimately causing organ damage. Current treatments often lack specificity, necessitating high doses, prolonged usage, and high recurrence rates. Therefore, the identification of innovative and safe therapeutic strategies is urgently required. Recent preclinical studies and clinical trials on inflammatory and autoimmune diseases have evidenced the immunosuppressive properties of mesenchymal stromal cells (MSCs). Studies have demonstrated that extracellular vesicles (EV) derived from MSCs can mitigate abnormal autoinflammation while maintaining safety within the diseased microenvironment. This study conducted a systematic review to elucidate the crucial role of MSC-EVs in alleviating autoimmune diseases, particularly focusing on their impact on the underlying mechanisms of autoimmune conditions such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). By specifically examining the regulatory functions of microRNAs (miRNAs) derived from MSC-EVs, the comprehensive study aimed to enhance the understanding related to disease mechanisms and identify potential diagnostic markers and therapeutic targets for these diseases.


Subject(s)
Autoimmune Diseases , Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Lupus Erythematosus, Systemic/therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Animals , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Immunomodulation
10.
J Cell Mol Med ; 28(10): e18445, 2024 May.
Article in English | MEDLINE | ID: mdl-38801403

ABSTRACT

Chronic prostatitis and chronic pelvic pain syndrome (CP/CPPS), a prevalent urological ailment, exerts a profound influence upon the well-being of the males. Autoimmunity driven by Th17 cells has been postulated as a potential factor in CP/CPPS pathogenesis. Nonetheless, elucidating the precise mechanisms governing Th17 cell recruitment to the prostate, triggering inflammation, remained an urgent inquiry. This study illuminated that CCL20 played a pivotal role in attracting Th17 cells to the prostate, thereby contributing to prostatitis development. Furthermore, it identified prostate stromal cells and immune cells as likely sources of CCL20. Additionally, this research unveiled that IL-17A, released by Th17 cells, could stimulate macrophages to produce CCL20 through the NF-κB/MAPK/PI3K pathway. The interplay between IL-17A and CCL20 establishes a positive feedback loop, which might serve as a critical mechanism underpinning the development of chronic prostatitis, thus adding complexity to its treatment challenges.


Subject(s)
Autoimmune Diseases , Chemokine CCL20 , Chemotaxis , Interleukin-17 , Prostatitis , Th17 Cells , Male , Prostatitis/immunology , Prostatitis/pathology , Prostatitis/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Chemokine CCL20/metabolism , Chemokine CCL20/genetics , Animals , Interleukin-17/metabolism , Interleukin-17/immunology , Mice , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal , NF-kappa B/metabolism , Signal Transduction , Humans , Mice, Inbred C57BL , Prostate/pathology , Prostate/metabolism , Prostate/immunology , Phosphatidylinositol 3-Kinases/metabolism , Autoimmunity
11.
Mod Pathol ; 37(6): 100491, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38588886

ABSTRACT

Patients with autoimmune gastritis (AIG) have a 13-fold risk of developing type-1 neuroendocrine tumors, whereas the risk for gastric adenocarcinoma is still uncertain. Here we describe the clinicopathologic and molecular features of a series of gastric carcinomas (GC) arising in the context of AIG. A total of 26 AIG-associated GC specimens were collected from 4 Italian Institutions. Immunohistochemistry for MUC1, MUC2, MUC5AC, MUC6, CDX2, HER2, PD-L1, CLDN18, mismatch repair (MMR) proteins, and p53 and EBV-encoded RNA (EBER) in situ hybridization were performed. Histologic and immunohistochemical features were jointly reviewed by 5 expert gastrointestinal pathologists. Next-generation sequencing analysis (TrueSight Oncology 500, Illumina) of 523 cancer-related genes was performed on 19 cases. Most tumors were diagnosed as pT1 (52%) and they were located in the corpus/fundus (58%) and associated with operative link for gastritis assessment stage II gastritis (80.8%), absence of parietal cells, complete intestinal metaplasia, and enterochromaffin-like-cell micronodular hyperplasia. Only 4 (15.4%) GCs were diagnosed during follow-up for AIG. The following histotypes were identified: 20 (77%) adenocarcinomas; 3 (11%) mixed neuroendocrine-non-neuroendocrine neoplasms, and 2 (8%) high-grade solid adenocarcinomas with focal neuroendocrine component, 1 (4%) adenocarcinoma with an amphicrine component. Overall, 7 cases (27%) showed MMR deficiency, 3 (12%) were positive (score 3+) for HER2, 6 (23%) were CLDN18 positive, and 11 (42%) had PD-L1 combined positive score ≥ 10. EBER was negative in all cases. Molecular analysis revealed 5/19 (26%) microsatellite instability (MSI) cases and 7 (37%) tumor mutational burden (TMB) high. The most frequently altered genes were TP53 (8/19, 42%), RNF43 (7/19, 37%), ERBB2 (7/19, 37% [2 amplified and 5 mutated cases]), ARID1A (6/19, 32%), and PIK3CA (4/19, 21%). In summary, AIG-associated GCs are often diagnosed at low stage in patients with longstanding misrecognized severe AIG; they often display a neuroendocrine component or differentiation, have relatively higher rates of MMR deficiency, and TMB high.


Subject(s)
Autoimmune Diseases , Gastritis , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Male , Female , Gastritis/pathology , Gastritis/genetics , Gastritis/immunology , Aged , Middle Aged , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Adult , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Aged, 80 and over
12.
Methods Mol Biol ; 2782: 159-166, 2024.
Article in English | MEDLINE | ID: mdl-38622400

ABSTRACT

Regulatory B (Breg) cells have been demonstrated to play an important role in the inhibition of a wide range of immunological responses, and they are absent or malfunction in autoimmune diseases like lupus. Breg cells can control immunological responses and keep the immune system in a balanced state by releasing immunosuppressive cytokines such as transforming growth factor-beta (TGF-ß) and interleukin-10 (IL-10), which in turn promote regulatory T (Treg) cells and reduce effector T cell responses. Breg cells have also been linked to the modulation of cancer immunity. Due to their immunosuppressive role, in the context of cancer, Breg cells aid in tumor immune evasion and promote tumor progression. Nonetheless, it has been established that Breg cells are involved in both cancer immunity and autoimmunity, and their characterizations beyond surface markers, for example, on the transcriptomic level, are essential for our understanding of Breg biology in health and disease. In this chapter, using lupus-prone MRL/lpr mice, we describe a Breg cell isolation protocol for the purpose of single-cell RNA sequencing analysis.


Subject(s)
Autoimmune Diseases , B-Lymphocytes, Regulatory , Neoplasms , Animals , Mice , Mice, Inbred MRL lpr , Cytokines/metabolism , Transforming Growth Factor beta/genetics , T-Lymphocytes, Regulatory , Autoimmune Diseases/pathology , Neoplasms/pathology
14.
Br J Dermatol ; 191(2): 261-266, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38581445

ABSTRACT

BACKGROUND: Artificial intelligence (AI) is reshaping healthcare, using machine and deep learning (DL) to enhance disease management. Dermatology has seen improved diagnostics, particularly in skin cancer detection, through the integration of AI. However, the potential of AI in automating immunofluorescence imaging for autoimmune bullous skin diseases (AIBDs) remains untapped. While direct immunofluorescence (DIF) supports diagnosis, its manual interpretation can hinder efficiency. The use of DL to classify DIF patterns automatically, including the intercellular (ICP) and linear pattern (LP), holds promise for improving the diagnosis of AIBDs. OBJECTIVES: To develop AI algorithms for automated classification of AIBD DIF patterns, such as ICP and LP, in order to enhance diagnostic accuracy, streamline disease management and improve patient outcomes through DL-driven immunofluorescence interpretation. METHODS: We collected immunofluorescence images from skin biopsies of patients suspected of having an AIBD between January 2022 and January 2024. Skin tissue was obtained via a 5-mm punch biopsy, prepared for DIF. Experienced dermatologists classified the images as ICP, LP or negative. To evaluate our DL approach, we divided the images into training (n = 436) and test sets (n = 93). We employed transfer learning with pretrained deep neural networks and conducted fivefold cross-validation to assess model performance. Our dataset's class imbalance was addressed using weighted loss and data augmentation strategies. The models were trained for 50 epochs using Pytorch, achieving an image size of 224 × 224 pixels for both convolutional neural networks (CNNs) and the Swin Transformer. RESULTS: Our study compared six CNNs and the Swin Transformer for AIBD image classification, with the Swin Transformer achieving the highest average validation accuracy (98.5%). On a separate test set, the best model attained an accuracy of 94.6%, demonstrating 95.3% sensitivity and 97.5% specificity across AIBD classes. Visualization with Grad-CAM (class activation mapping) highlighted the model's reliance on characteristic patterns for accurate classification. CONCLUSIONS: The study highlighted the accuracy of CNNs in identifying DIF features. This approach aids automated analysis and reporting, offering reproducibility, speed, data handling and cost-efficiency. Integrating DL into skin immunofluorescence promises precise diagnostics and streamlined reporting in this branch of dermatology.


Artificial intelligence (AI) is transforming healthcare through machine and deep learning (computer systems that can learn and adapt, and make complex decisions, without receiving explicit instructions), improving disease management in dermatology, particularly in detecting skin cancer. However, AI's potential in automating immunofluorescence imaging in autoimmune bullous (blistering) skin diseases (AIBDs) remains largely untapped. Manual interpretation of direct immunofluorescence (DIF ­ a type of microscopy) can reduce efficiency. However, using deep learning to automatically classify DIF patterns (for example, the 'intercellular pattern' (ICP) and the 'linear pattern' (LP)) holds promise in helping with the diagnosis of AIBDs. This study aimed to develop AI algorithms for the automated classification of AIBD DIF patterns, such as ICP and LP, to improve diagnostic accuracy and streamline disease management. Immunofluorescence images were collected from skin biopsies of patients with a suspected AIBD between January 2022 and January 2024. Dermatologists classified the images into three categories: ICP, LP and negative. The dataset was divided into training (436 images) and test sets (93 images). A transfer learning framework (where what has been learned previously in one setting is used to improve performance in another) was used to make up for the limited amount of training data, to explore different models for the AIBD classification task. Our results revealed that a model called the 'Swin Transformer' achieved an average accuracy of 99% in diagnosing different AIBDs. The best model attained 95% accuracy on the test set and was reliable in identifying and ruling out different AIBDs. Visualization with Grad-CAM (a technique used in deep learning) highlighted the model's use of characteristic patterns to classify the diseases accurately. Overall, integrating deep learning in skin immunofluorescence promises to improve diagnostics and streamline reporting in dermatology, which could improve consistency, speed and cost-efficiency.


Subject(s)
Autoimmune Diseases , Deep Learning , Skin Diseases, Vesiculobullous , Humans , Skin Diseases, Vesiculobullous/diagnosis , Skin Diseases, Vesiculobullous/pathology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Fluorescent Antibody Technique, Direct/methods , Skin/pathology , Skin/immunology , Biopsy , Algorithms
15.
Biochimie ; 223: 54-73, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38657832

ABSTRACT

Mesenchymal Stem Cells (MSCs) are of interest in the clinic because of their immunomodulation capabilities, capacity to act upstream of inflammation, and ability to sense metabolic environments. In standard physiologic conditions, they play a role in maintaining the homeostasis of tissues and organs; however, there is evidence that they can contribute to some autoimmune diseases. Gaining a deeper understanding of the factors that transition MSCs from their physiological function to a pathological role in their native environment, and elucidating mechanisms that reduce their therapeutic relevance in regenerative medicine, is essential. We conducted a Systematic Review and Meta-Analysis of human MSCs in preclinical studies of autoimmune disease, evaluating 60 studies that included 845 patient samples and 571 control samples. MSCs from any tissue source were included, and the study was limited to four autoimmune diseases: multiple sclerosis, rheumatoid arthritis, systemic sclerosis, and lupus. We developed a novel Risk of Bias tool to determine study quality for in vitro studies. Using the International Society for Cell & Gene Therapy's criteria to define an MSC, most studies reported no difference in morphology, adhesion, cell surface markers, or differentiation into bone, fat, or cartilage when comparing control and autoimmune MSCs. However, there were reported differences in proliferation. Additionally, 308 biomolecules were differentially expressed, and the abilities to migrate, invade, and form capillaries were decreased. The findings from this study could help to explain the pathogenic mechanisms of autoimmune disease and potentially lead to improved MSC-based therapeutic applications.


Subject(s)
Autoimmune Diseases , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Humans , Autoimmune Diseases/therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Differentiation , Animals , Mesenchymal Stem Cell Transplantation , Arthritis, Rheumatoid/therapy , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167198, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670439

ABSTRACT

Autoimmune inner ear disease (AIED) is an organ-specific disease characterized by irreversible, prolonged, and progressive hearing and equilibrium dysfunctions. The primary symptoms of AIED include asymmetric sensorineural hearing loss accompanied by vertigo, aural fullness, and tinnitus. AIED is divided into primary and secondary types. Research has been conducted using animal models of rheumatoid arthritis (RA), a cause of secondary AIED. However, current models are insufficient to accurately analyze vestibular function, and the mechanism underlying the onset of AIED has not yet been fully elucidated. Elucidation of the mechanism of AIED onset is urgently needed to develop effective treatments. In the present study, we analyzed the pathogenesis of vertigo in autoimmune diseases using a mouse model of type II collagen-induced RA. Auditory brain stem response analysis demonstrated that the RA mouse models exhibited hearing loss, which is the primary symptom of AIED. In addition, our vestibulo-oculomotor reflex analysis, which is an excellent vestibular function test, accurately captured vertigo symptoms in the RA mouse models. Moreover, our results revealed that the cause of hearing loss and vestibular dysfunction was not endolymphatic hydrops, but rather structural destruction of the organ of Corti and the lateral semicircular canal ampulla due to an autoimmune reaction against type II collagen. Overall, we were able to establish a mouse model of AIED without endolymphatic hydrops. Our findings will help elucidate the mechanisms of hearing loss and vertigo associated with AIED and facilitate the development of new therapeutic methods.


Subject(s)
Autoimmune Diseases , Disease Models, Animal , Endolymphatic Hydrops , Labyrinth Diseases , Animals , Mice , Endolymphatic Hydrops/pathology , Endolymphatic Hydrops/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/immunology , Labyrinth Diseases/pathology , Labyrinth Diseases/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/complications , Vertigo/pathology , Vertigo/etiology , Collagen Type II/immunology , Evoked Potentials, Auditory, Brain Stem , Female , Mice, Inbred C57BL
19.
Cell Rep ; 43(3): 113872, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38427562

ABSTRACT

Infection, autoimmunity, and cancer are principal human health challenges of the 21st century. Often regarded as distinct ends of the immunological spectrum, recent studies hint at potential overlap between these diseases. For example, inflammation can be pathogenic in infection and autoimmunity. T resident memory (TRM) cells can be beneficial in infection and cancer. However, these findings are limited by size and scope; exact immunological factors shared across diseases remain elusive. Here, we integrate large-scale deeply clinically and biologically phenotyped human cohorts of 526 patients with infection, 162 with lupus, and 11,180 with cancer. We identify an NKG2A+ immune bias as associative with protection against disease severity, mortality, and autoimmune/post-acute chronic disease. We reveal that NKG2A+ CD8+ T cells correlate with reduced inflammation and increased humoral immunity and that they resemble TRM cells. Our results suggest NKG2A+ biases as a cross-disease factor of protection, supporting suggestions of immunological overlap between infection, autoimmunity, and cancer.


Subject(s)
Autoimmune Diseases , Communicable Diseases , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Neoplasms/pathology , Autoimmunity , Inflammation/pathology , Autoimmune Diseases/pathology , Communicable Diseases/pathology , Immunologic Memory
20.
Hum Pathol ; 146: 28-34, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38518977

ABSTRACT

Lymphocytic lobulitis (LL) is characterized by prominent lymphocytic infiltrates centered on lobules. Sclerosing lymphocytic lobulitis (SCLL) associated with diabetes mellitus (DM) or autoimmune disease (AI) was the first type to be described. Subsequently, non-sclerosing LL (NSCLL) was reported as an incidental finding in prophylactic mastectomies due to high risk germline mutations or a family history of breast cancer. The two types of LL were distinguished by stromal features and a predominant population of B-cells in the former and T-cells in the latter. In this study, 8 cases of NSCLL detected clinically or by screening were compared to 44 cases of SCLL. One case of NSCLL presented as a palpable mass, 2 as masses on screening, and 5 as MRI enhancement. In contrast, 80% of SCLL cases presented as palpable masses. Half the cases of NSCLL were associated with a BRCA1 or 2 mutation compared to 1 case of SCLL (2%). Three additional cases of NSCLL were associated with a strong family and/or personal history of breast cancer. Almost half (52%) of SCLL cases were associated with DM or AI, but only 25% of NSCLL. Immunoperoxidase studies confirmed a predominance of T-cells in NSCLL and B-cells in SCLL associated with DM or AI. It is important for pathologists to be aware of this new observation that NSCLL can be detected as a palpable mass or an imaging finding in diagnostic biopsies, as its presence can be indicative of a significant risk for breast cancer.


Subject(s)
B-Lymphocytes , Breast Neoplasms , T-Lymphocytes , Humans , Female , Middle Aged , Adult , B-Lymphocytes/pathology , Biopsy , T-Lymphocytes/pathology , T-Lymphocytes/immunology , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Aged , Sclerosis , Breast/pathology , Breast/diagnostic imaging , Magnetic Resonance Imaging , Genetic Predisposition to Disease , Mutation , Autoimmune Diseases/pathology , Breast Diseases/pathology , Breast Diseases/diagnosis , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Mammography , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL