Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (6,881)

Search Parameters:
Keywords = autoimmune disease

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 357 KB  
Article
Interstitial Pneumonia with Autoimmune Features from the Rheumatologists’ Perspective; Single Center Experience
by Emine Uslu, Didem Sahin, Ahmet Ilbay, Recep Yilmaz, Abdulbaki Gaydan, Nilgun Govec Giynas, Ahmet Usta, Yeter Mahmutoglu, Rahime Aksoy, Serdar Sezer, Mucteba Enes Yayla, Melahat Kul, Aysegul Gursoy Coruh, Caglar Uzun, Ebru Us, Ozlem Ozdemir Kumbasar, Askin Ates and Tahsin Murat Turgay
Diagnostics 2026, 16(2), 299; https://doi.org/10.3390/diagnostics16020299 (registering DOI) - 17 Jan 2026
Abstract
Background/Objectives: Interstitial pneumonia with autoimmune features (IPAF) is a recently defined entity characterized by interstitial lung disease (ILD) with clinical, serological, and radiological features suggestive of autoimmunity that do not fulfil the criteria for a defined connective tissue disease (CTD). This study [...] Read more.
Background/Objectives: Interstitial pneumonia with autoimmune features (IPAF) is a recently defined entity characterized by interstitial lung disease (ILD) with clinical, serological, and radiological features suggestive of autoimmunity that do not fulfil the criteria for a defined connective tissue disease (CTD). This study aimed to evaluate the clinical characteristics, treatment modalities, and outcomes of patients with IPAF in a tertiary referral center. Methods: We retrospectively analyzed 72 patients who fulfilled the IPAF classification criteria. Demographic, clinical, serological, radiological, pulmonary function, treatment, and survival data were collected and evaluated. Logistic regression analysis was performed to identify factors associated with mortality. Results: The cohort consisted of 62.5% female patients, with a mean age of 62.7 (SD, 10.4) years at diagnosis. The most frequent radiological pattern was nonspecific interstitial pneumonia (83.3%). Raynaud’s phenomenon (6.9%) and arthritis (2.8%) were the most common rheumatological manifestations. Antinuclear antibodies positivity at titers ≥1:320 was observed in 27.8% of patients. Azathioprine was the most frequently prescribed agent (20.8%), followed by mycophenolate mofetil (11.1%). After a median follow-up of 30.1 months (IQR, 52.8), 16 patients (22.22%) died, with a 5-year survival rate of 70%. Glucocorticoid therapy at doses ≥20 mg/day was independently associated with increased mortality (OR 6.13 (95% CI 1.17–32.21). Conclusions: IPAF predominantly affects middle-aged females. Glucocorticoid use at doses ≥20 mg/day was associated with mortality; however, this observational association may reflect underlying disease severity rather than a causal effect of high-dose treatment. Further prospective studies are needed to optimize management strategies in patients with IPAF. Full article
(This article belongs to the Section Clinical Diagnosis and Prognosis)
Show Figures

Figure 1

10 pages, 410 KB  
Article
Accommodating Celiac Disease in Higher Education: Evidence-Informed National Recommendations
by Vanessa Weisbrod, Meghan Donnelly McKeon, Emma Kowzun, Marilyn Grunzweig Geller, Jackie Jossen, Marisa Gallant Stahl, Maureen M. Leonard, Mary Shull, Janis Arnold, Jennifer Kumin, Sharon Weston, Anne R. Lee, Mary Vargas, Dale Lee, Allyson West, Catherine Raber, Katherine Vera Sachs and Ritu Verma
Nutrients 2026, 18(2), 294; https://doi.org/10.3390/nu18020294 (registering DOI) - 16 Jan 2026
Abstract
Objectives: We aimed to develop expert-informed recommendations for colleges and universities to support students with celiac disease (CeD) managing a gluten-free (GF) diet. Methods: A multidisciplinary panel of 40 stakeholders, including physicians, dietitians, a disability rights attorney, university staff, and students, was convened [...] Read more.
Objectives: We aimed to develop expert-informed recommendations for colleges and universities to support students with celiac disease (CeD) managing a gluten-free (GF) diet. Methods: A multidisciplinary panel of 40 stakeholders, including physicians, dietitians, a disability rights attorney, university staff, and students, was convened by the Celiac Disease Foundation to create expert-based and experience-informed recommendations. Over a 6-month period, the group conducted literature reviews, stakeholder interviews, and expert consensus discussions to identify common barriers and accommodations aligned with federal disability law. The expert panel collaboratively developed and revised an initial set of recommendations. Two rounds of structured voting were held during which panelists provided feedback to refine content and ensure clarity. All final recommendations were adopted with at least 90% of panelists voting in support. Results: The panel identified 24 accommodations across four domains: academics, housing, dining, and campus life. Academic recommendations include flexibility for illness-related absences, support for remote learning, and classroom modifications. Housing recommendations emphasize access to priority placement, appropriate appliances, and proximity to safe dining. Dining accommodations address GF food availability, ingredient transparency, staff training, and meal plan flexibility. Campus life recommendations ensure full participation in athletics, study abroad, social events, and internships, with supports for psychosocial well-being. Conclusions: This manuscript presents the first expert-informed recommendations focused specifically on the needs of college students with CeD. These recommendations are intended to support institutions as they develop strategies to enhance access to GF food, quality of life, educational supports, and student experience for those living with this chronic autoimmune condition. Full article
(This article belongs to the Special Issue The Implications of Celiac Disease and the GFD on Health Outcomes)
23 pages, 1051 KB  
Review
Early-Life Gut Microbiota: Education of the Immune System and Links to Autoimmune Diseases
by Pleun de Groen, Samantha C. Gouw, Nordin M. J. Hanssen, Max Nieuwdorp and Elena Rampanelli
Microorganisms 2026, 14(1), 210; https://doi.org/10.3390/microorganisms14010210 - 16 Jan 2026
Abstract
Early life is a critical window for immune system development, during which the gut microbiome shapes innate immunity, antigen presentation, and adaptive immune maturation. Disruptions in microbial colonization—driven by factors such as cesarean delivery, antibiotic exposure, and formula feeding—deplete beneficial early-life taxa (e.g., [...] Read more.
Early life is a critical window for immune system development, during which the gut microbiome shapes innate immunity, antigen presentation, and adaptive immune maturation. Disruptions in microbial colonization—driven by factors such as cesarean delivery, antibiotic exposure, and formula feeding—deplete beneficial early-life taxa (e.g., Bifidobacterium, Bacteroides, and Enterococcus) and impair key microbial functions, including short-chain fatty acid (SCFA) production by these keystone species, alongside regulatory T cell induction. These dysbiosis patterns are associated with an increased risk of pediatric autoimmune diseases, notably type 1 diabetes, inflammatory bowel disease, celiac disease, and juvenile idiopathic arthritis. This review synthesizes current evidence on how the early-life microbiota influences immune maturation, with potential effects on the development of autoimmune diseases later in life. We specifically focus on human observational and intervention studies, where treatments with probiotics, synbiotics, vaginal microbial transfer, or maternal fecal microbiota transplantations have been shown to partially restore a disrupted microbiome. While restoration of the gut microbiome composition and function is the main reported outcome of these studies, to date, no reports have disclosed direct prevention of autoimmune disease development by targeting the early-life gut microbiome. In this regard, a better understanding of the early-life microbiome–immune axis is essential for developing targeted preventive strategies. Future research must prioritize longitudinal evaluation of autoimmune outcomes after microbiome modulation to reduce the burden of chronic immune-mediated diseases. Full article
(This article belongs to the Special Issue Microbiomes in Human Health and Diseases)
32 pages, 3412 KB  
Review
Engineering Immunity: Current Progress and Future Directions of CAR-T Cell Therapy
by Mouldy Sioud and Nicholas Paul Casey
Int. J. Mol. Sci. 2026, 27(2), 909; https://doi.org/10.3390/ijms27020909 - 16 Jan 2026
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative form of immunotherapy, enabling the precise engineering of T cells to recognize and eliminate pathogenic cells. In hematologic malignancies, CAR-T cells targeting CD19 or B cell maturation antigens have achieved remarkable remission [...] Read more.
Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative form of immunotherapy, enabling the precise engineering of T cells to recognize and eliminate pathogenic cells. In hematologic malignancies, CAR-T cells targeting CD19 or B cell maturation antigens have achieved remarkable remission rates and durable responses in patients with otherwise refractory disease. Despite these successes, extending CAR-T cell therapy to solid tumors remains challenging due to antigen heterogeneity, poor T cell infiltration, and the immunosuppressive tumor microenvironment (TME). Beyond oncology, CAR-T cell therapy has also shown promise in autoimmune diseases, where early clinical studies suggest that B cell-directed CAR-T cells can induce sustained remission in conditions such as systemic lupus erythematosus. This review highlights advances in CAR-T cell engineering, including DNA- and mRNA-based platforms for ex vivo and in vivo programming, and discusses emerging strategies to enhance CAR-T cell trafficking, persistence, and resistance to TME. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Immunotherapy in Cancer)
Show Figures

Figure 1

11 pages, 701 KB  
Article
Vitamin D Deficiency and Replacement Challenges in Type 1 Gastric Neuroendocrine Tumors: A Comparative Study
by Elio Benevento, Michele Coletta, Alessia Liccardi, Roberto Minotta, Gianfranco Di Iasi, Massimo Di Nola, Annamaria Colao and Roberta Modica
Nutrients 2026, 18(2), 281; https://doi.org/10.3390/nu18020281 - 15 Jan 2026
Abstract
Background/Objectives: Type 1 gastric neuroendocrine tumors (gNET) arise in the setting of autoimmune chronic atrophic gastritis and secondary hypergastrinemia. Vitamin D deficiency (VDD) has been associated with bone impairment and adverse outcomes in patients with neuroendocrine tumor (NET); however, data specifically addressing [...] Read more.
Background/Objectives: Type 1 gastric neuroendocrine tumors (gNET) arise in the setting of autoimmune chronic atrophic gastritis and secondary hypergastrinemia. Vitamin D deficiency (VDD) has been associated with bone impairment and adverse outcomes in patients with neuroendocrine tumor (NET); however, data specifically addressing gNET remain limited. This study aimed to evaluate vitamin D status, supplementation requirements, and bone involvement in patients with type 1 gNET compared with those with entero-pancreatic NET (EP-NET). Methods: This retrospective study included patients with type 1 gNET followed at a tertiary referral center between 2010 and 2025 and an age- and sex-matched EP-NET cohort. VDD prevalence, time and dose required for normalization, supplementation formulations, bone status, and dietary habits were analyzed. Results: Twenty-six patients were included (thirteen gNET and thirteen EP-NET). VDD was significantly more prevalent in the gNET group compared with the EP-NET group (92.3% vs. 46.2%, p = 0.03, OR: 14). gNET required significantly higher daily cholecalciferol doses (3198.9 ± 1629 vs. 1580 ± 1121 IU/day, p = 0.008) and more frequently required multiple supplementation formulations (38.5% vs. 0%, p = 0.04). Multivariable linear regression analysis restricted to VDD patients confirmed that gNET was independently associated with higher daily cholecalciferol dose requirements (p = 0.037). Bone impairment, defined as osteoporosis or osteopenia, was significantly more common in the gNET group (61.5% vs. 15.4%, p = 0.04, OR: 8.8). Dietary adherence did not differ between groups. Conclusions: Type 1 gNET show a higher burden of VDD, increased vitamin D supplementation requirements, and a higher prevalence of bone impairment compared with EP-NET, irrespective of dietary habits. These findings suggest disease-specific mechanisms and support the need for tailored management in these patients. Full article
Show Figures

Figure 1

9 pages, 269 KB  
Article
Asymmetric Tear Secretion: Can This Disorder Help in Suspecting Gastroesophageal Reflux Disease and in Managing Sjögren’s Disease? A Pilot Study
by Vilius Kontenis, Jūratė Gruodė, Jurgita Urbonienė, Almantas Šiaurys and Diana Mieliauskaitė
Medicina 2026, 62(1), 176; https://doi.org/10.3390/medicina62010176 - 15 Jan 2026
Abstract
Background and Objectives: Patients with Sjögren’s disease (SjD) do not experience any improvement in gastroesophageal reflux disease (GERD) symptoms after SjD treatment, and in some patients, reflux even worsens. It is important to note that GERD manifests itself through typical and atypical [...] Read more.
Background and Objectives: Patients with Sjögren’s disease (SjD) do not experience any improvement in gastroesophageal reflux disease (GERD) symptoms after SjD treatment, and in some patients, reflux even worsens. It is important to note that GERD manifests itself through typical and atypical symptoms, the latter of which may include eye damage, as evidenced by a growing body of research. When SjD patients were prescribed medication to treat GERD, their condition improved at the same time. Therefore, we aim to investigate whether there is a link between ocular dryness and gastroesophageal reflux disease (GERD) in patients with Sjögren’s disease (SjD). Materials and Methods: Our study included 27 patients with SjD according to the 2016 American College of Rheumatology and the European League Against Rheumatism (ACR/EULAR) Sjögren’s syndrome Classification Criteria, and 28 patients with non-autoimmune sicca syndrome due to GERD (nonautoimmSicca). Results: The study involved 55 participants, 48 (87.3%) women and 7 (12.7%) men. The median age was 54 years (IQR 49–64). A total of 41 subjects (74.5%) had GERD, and 20 subjects (36.4%) tested positive for Helicobacter pylori: 13 (48.1%) and 1 (3.7%) in the SjD group, and 28 (100.0%) and 19 (67.9%) in the nonautoimmSicca group, respectively. A significant difference in asymmetric tear secretion (p < 0.001) was found between the nonautoimmSicca and SjD patients, with values of 5 (3–10) mm/5 min and 1 (0–2) mm/5 min, respectively. A low correlation was detected between sialometry results and tear secretion asymmetry (r = 0.48, p < 0.001). An increase of 1 mm/5 min in the tear secretion asymmetry between the eyes was associated with a 2.04-fold increase in the odds ratio for having GERD (95% CI 1.25–3.32, p = 0.004), and was associated with a 1.9-fold increase in the odds ratio for having GERD (95% CI 1.04–3.49, p = 0.038) in patients with SjD. The presence of Helicobacter pylori is associated with asymmetric tear secretion [95% CI 1.22 (1.05–1.41, p = 0.010)]. Conclusions: Asymmetric tear secretion between the eyes is associated with the odds of having GERD. Patients with non-autoimmune sicca syndrome due to GERD have significantly greater asymmetry in tear secretion compared to those diagnosed with Sjögren’s disease. Full article
(This article belongs to the Special Issue Recent Advances in Autoimmune Rheumatic Diseases—3rd Edition)
18 pages, 3360 KB  
Article
ZechariahNet: A Novel Method of MS Lesion Diagnosis Through MRI Images by the Combination of C-LSTM and 3D CNN Algorithms
by Mahshid Dehghanpour, Mansoor Fateh, Zeynab Mohammadpoory and Saideh Ferdowsi
Algorithms 2026, 19(1), 72; https://doi.org/10.3390/a19010072 - 15 Jan 2026
Abstract
In light of the growing prevalence of the autoimmune disease multiple sclerosis (MS), accurate detection of MS lesions in brain magnetic resonance imaging (MRI) images plays a critical role in assisting neurologists with timely diagnosis. The high similarity between MS lesions and normal [...] Read more.
In light of the growing prevalence of the autoimmune disease multiple sclerosis (MS), accurate detection of MS lesions in brain magnetic resonance imaging (MRI) images plays a critical role in assisting neurologists with timely diagnosis. The high similarity between MS lesions and normal brain tissues, however, makes this task particularly challenging. Although numerous deep-learning-based approaches have been proposed for the automatic segmentation of MS lesions, the method presented in this study has achieved superior results. ZechariahNet is a U-Net-based architecture that integrates transition down blocks, squeeze-attention (SA) blocks, dense blocks, and Convolutional LSTM (C-LSTM) blocks within a 3D CNN framework. By jointly exploiting spatial–temporal information from three consecutive MRI slices (previous, current, and subsequent) and strategically applying C-LSTM modules across the encoder and decoder paths, the proposed model effectively captures the neighborhood dependencies for enhanced feature extraction and reconstruction. These architectural innovations significantly improve segmentation accuracy, enabling ZechariahNet to achieve a dice similarity coefficient (DSC) of 84.72%, outperforming existing state-of-the-art methods. Full article
Show Figures

Figure 1

19 pages, 3649 KB  
Review
TIM-3+ Macrophages: Insights into Their Role in Cancer and Inflammation
by Aleksandra Maksimova, Tamara Tyrinova and Elena Chernykh
Int. J. Mol. Sci. 2026, 27(2), 840; https://doi.org/10.3390/ijms27020840 - 14 Jan 2026
Viewed by 26
Abstract
T-cell immunoglobulin and mucin domain 3 (TIM-3), a well-known immune checkpoint molecule, is increasingly recognized for its regulatory functions beyond T cell exhaustion, particularly in macrophages. Recent advances have revealed the important role of this molecule in various pathological and physiological conditions. The [...] Read more.
T-cell immunoglobulin and mucin domain 3 (TIM-3), a well-known immune checkpoint molecule, is increasingly recognized for its regulatory functions beyond T cell exhaustion, particularly in macrophages. Recent advances have revealed the important role of this molecule in various pathological and physiological conditions. The demand for a comprehensive study of TIM-3 is increasing, particularly as a result of ongoing clinical trials targeting TIM-3 in oncology. This review is devoted to the role of TIM-3 in macrophage biology, focusing on associations between TIM-3 expression and macrophage polarization states and functional activity, as well as its involvement in the pathogenesis of different diseases and reproductive immunology. The review examines known effects and molecular mechanisms by which TIM-3 regulates macrophage functional phenotype and the contribution of TIM-3-expressing macrophages to cancer, pregnancy, inflammation, infectious and autoimmune diseases, and fibrosis. Findings highlight the controversial role of TIM-3 in the regulatory function of macrophages and suggest that TIM-3 functions differently depending on the context. The review also touches on gaps and unexplored parts of the topic. A summary of current data allows us to conclude that TIM-3 is an important modulator of macrophage functions and can be considered a potential therapeutic target in various pathological conditions. Full article
(This article belongs to the Special Issue The Role of Macrophages in Inflammation and Cancer: An Update)
Show Figures

Graphical abstract

17 pages, 2010 KB  
Article
Molecular Mimicry Between Trypanosoma cruzi and Human TUBB as a Potential Autoimmune Mechanism in Chagas
by Ana Valentina Centeno-Iglesias, Celeste Abigail Quille-Juarez, Paul Galvez-Murillo, Anggie Stefany Revilla-Zeballos, Gustavo Alberto Obando-Pereda and Luis Alberto Ponce-Soto
Immuno 2026, 6(1), 8; https://doi.org/10.3390/immuno6010008 - 14 Jan 2026
Viewed by 125
Abstract
Chagas disease, caused by Trypanosoma cruzi, affects a significant proportion of patients who develop digestive and cardiac complications, including megaviscera. This pathogenesis has been associated with autoimmune mechanisms mediated by molecular mimicry. In this study, an in silico evaluation of the potential [...] Read more.
Chagas disease, caused by Trypanosoma cruzi, affects a significant proportion of patients who develop digestive and cardiac complications, including megaviscera. This pathogenesis has been associated with autoimmune mechanisms mediated by molecular mimicry. In this study, an in silico evaluation of the potential structural basis of cross-reactivity of β-tubulin 1.9 of T. cruzi and the human β-4A tubulin isoform 3 was conducted. Using bioinformatics tools, homologous regions were identified and potentially immunogenic epitopes were predicted, considering their structural modeling and molecular docking. The proteins shared 87% sequence identity and 95% similarity, with an almost identical structural overlap, RMSD 0.291 Å. Three epitopes, VPFPRLHFF, NDLVSEYQQYQDATI, and GQSGAGNNWAKGHYTEGAELIDS, exhibited high predicted antigenicity, with the 9-mer and 16-mer peptides displaying structurally compatible docking poses within the binding grooves of MHC class I and class II molecules, respectively, while B-cell epitope potential was inferred from sequence-based property predictions. Normal mode analysis, used as an exploratory approach, suggested comparable flexibility profiles for the parasitic- and human-derived peptide–MHC complexes. These findings provide an exploratory structural framework supporting a potential role of β-tubulin epitopes in molecular mimicry processes implicated in the development of chagasic megaviscera. Full article
(This article belongs to the Section Autoimmunity and Immunoregulation)
Show Figures

Figure 1

41 pages, 4351 KB  
Review
Autoantibodies as Precision Tools in Connective Tissue Diseases: From Epiphenomenon to Endophenotype
by Muhammad Soyfoo and Julie Sarrand
Antibodies 2026, 15(1), 7; https://doi.org/10.3390/antib15010007 - 13 Jan 2026
Viewed by 69
Abstract
Autoantibodies have long been regarded as passive reflections of immune dysregulation in connective tissue diseases (CTDs). Recent advances in systems immunology and molecular pathology have fundamentally redefined them as active molecular fingerprints that delineate distinct disease endophenotypes with predictive power for clinical trajectories [...] Read more.
Autoantibodies have long been regarded as passive reflections of immune dysregulation in connective tissue diseases (CTDs). Recent advances in systems immunology and molecular pathology have fundamentally redefined them as active molecular fingerprints that delineate distinct disease endophenotypes with predictive power for clinical trajectories and therapeutic responses. Rather than mere epiphenomena, autoantibodies encode precise information about dominant immune pathways, organ tropism, and pathogenic mechanisms. This review synthesizes emerging evidence that autoantibody repertoires—defined by specificity, structural properties, and functional characteristics—stratify patients beyond traditional clinical taxonomy into discrete pathobiological subsets. Specific signatures such as anti-MDA5 in rapidly progressive interstitial lung disease, anti-RNA polymerase III in scleroderma renal crisis, and anti-Ro52/TRIM21 in systemic overlap syndromes illustrate how serological profiles predict outcomes with remarkable precision. Mechanistically, autoantibody pathogenicity is modulated by immunoglobulin isotype distribution, Fc glycosylation patterns, and tissue-specific receptor expression—variables that determine whether an antibody functions as a biomarker or pathogenic effector. The structural heterogeneity of autoantibodies, shaped by cytokine microenvironments and B-cell subset imprinting, creates a dynamic continuum between pro-inflammatory and regulatory states. The integration of serological, transcriptomic, and imaging data establishes a precision medicine framework: autoantibodies function simultaneously as disease classifiers and therapeutic guides. This endophenotype-driven approach is already influencing trial design and patient stratification in systemic lupus erythematosus, systemic sclerosis, and inflammatory myopathies, and is reshaping both clinical practice and scientific taxonomy in CTDs. Recognizing autoantibodies as endophenotypic determinants aligns disease classification with pathogenic mechanism and supports the transition towards immunologically informed therapeutic strategies. Full article
(This article belongs to the Special Issue Antibody and Autoantibody Specificities in Autoimmunity)
Show Figures

Graphical abstract

22 pages, 1508 KB  
Article
m6A-Modified Nucleotide Bases Improve Translation of In Vitro-Transcribed Chimeric Antigen Receptor (CAR) mRNA in T Cells
by Nga Lao, Simeng Li, Marina Ainciburu and Niall Barron
Int. J. Mol. Sci. 2026, 27(2), 796; https://doi.org/10.3390/ijms27020796 - 13 Jan 2026
Viewed by 66
Abstract
Lentiviral transduction remains the gold standard in adoptive modified cellular therapy, such as CAR-T; however, genome integration is not always desirable, such as when treating non-fatal autoimmune disease or for additional editing steps using CRISPR to produce allogeneic CAR-modified cells. Delivering in vitro-transcribed [...] Read more.
Lentiviral transduction remains the gold standard in adoptive modified cellular therapy, such as CAR-T; however, genome integration is not always desirable, such as when treating non-fatal autoimmune disease or for additional editing steps using CRISPR to produce allogeneic CAR-modified cells. Delivering in vitro-transcribed (IVT) mRNA represents an alternative solution but the labile nature of mRNA has led to efforts to improve half-life and translation efficiencies using a range of approaches including chemical and structural modifications. In this study, we explore the role of N6–methyladenosine (m6A) in a CD19-CAR sequence when delivered to T cells as an IVT mRNA. In silico analysis predicted the presence of four m6A consensus (DRACH) motifs in the CAR coding sequence and treating T cells with an inhibitor of the m6A methyltransferase (METTL3) resulted in a significant reduction in CAR protein expression. RNA analysis confirmed m6A bases at three of the predicted sites, indicating that the modification occurs independently of nuclear transcription. Synonymous mutation of the DRACH sites reduced the levels of CAR protein from 15 to >50% depending on the T cell donor. We also tested a panel of CAR transcripts with different UTRs, some containing m6A consensus motifs, and identified those which further improved protein expression. Furthermore, we found that the methylation of consensus m6A sites seems to be somewhat sequence-context-dependent. These findings demonstrate the importance of the m6A modification in stabilising and enhancing expression from IVT-derived mRNA and that this occurs within the cell, meaning targeted in vitro chemical modification during mRNA manufacturing may not be necessary. Full article
(This article belongs to the Collection Feature Papers in “Molecular Biology”)
Show Figures

Figure 1

37 pages, 6099 KB  
Review
Is Obesity a Modifiable Risk Factor in Multiple Sclerosis? Mechanistic Insights into Neuroinflammation and Oxidative Damage
by Fani-Niki Varra, Olga Pagonopoulou, Michail Varras, Viktoria-Konstantina Varra and Panagiotis Theodosis-Nobelos
Pathophysiology 2026, 33(1), 5; https://doi.org/10.3390/pathophysiology33010005 - 13 Jan 2026
Viewed by 64
Abstract
Introduction: Multiple sclerosis (MS) is a chronic autoimmune inflammatory disorder of the central nervous system (CNS) that leads to demyelination of CNS neurons and is influenced by genetic, environmental, and lifestyle factors, including diet and obesity. Methods: This review aims to [...] Read more.
Introduction: Multiple sclerosis (MS) is a chronic autoimmune inflammatory disorder of the central nervous system (CNS) that leads to demyelination of CNS neurons and is influenced by genetic, environmental, and lifestyle factors, including diet and obesity. Methods: This review aims to analyze at the molecular level the relationship between obesity, as a chronic inflammatory condition, and the pathophysiology of MS, as a chronic autoimmune inflammatory disease, in order to understand the complex links between obesity and MS through a search of the PubMed and Google Scholar databases. Discussion: Chronic inflammation and OS are interconnected processes, causing a toxic state, which contributes to the development of CNS neuroinflammation and neuronal damage, resulting in neuronal demyelination and the onset of MS. Adipose tissue is a complex endocrine organ; in addition to being a lipid storage organ, it secretes cytokines and adipokines, which are involved in the regulation of hormones, metabolism, inflammation, and whole-body homeostasis. Obesity triggers chronic low-grade inflammation, disruption of the blood–brain barrier (BBB) and brain metabolism, infiltration of the CNS by immune cells, production of ROS, and generation of oxidative stress (OS). Anti-inflammatory and pro-inflammatory adipokines are also implicated in MS and obesity. Conclusions: Obesity affects MS through common underlying mechanisms and seems to be a modifiable risk factor. Antioxidant and anti-inflammatory compounds with multi-functional characteristics could be additional tools to slow the progression of MS and its promotion through obesity while also offering potential treatment options for both conditions via their multi-targeting characteristics. Full article
Show Figures

Graphical abstract

30 pages, 1723 KB  
Article
Candidate Interaction Partners of Calpain-5 Suggest Clues to Its Involvement in Neovascular Inflammatory Vitreoretinopathy
by Jozsef Gal, Vimala Bondada, Rachel Crasta, Dorothy E. Croall, Calvin P. Vary and James W. Geddes
Cells 2026, 15(2), 142; https://doi.org/10.3390/cells15020142 - 13 Jan 2026
Viewed by 96
Abstract
Although calpain-5/CAPN5 is widely expressed in mammals, little is known regarding its functions. Pathogenic mutations of CAPN5 are causal for a devastating autoimmune eye disease, neovascular inflammatory vitreoretinopathy (NIV). To provide insight into both the physiological and pathological roles of CAPN5, it is [...] Read more.
Although calpain-5/CAPN5 is widely expressed in mammals, little is known regarding its functions. Pathogenic mutations of CAPN5 are causal for a devastating autoimmune eye disease, neovascular inflammatory vitreoretinopathy (NIV). To provide insight into both the physiological and pathological roles of CAPN5, it is essential to identify candidate interaction partners and possible substrates. Human SH-SY5Y neuroblastoma cells, transfected with full-length catalytically dead (Cys81Ala) CAPN5-3×FLAG, were used for anti-FLAG co-immunoprecipitation (co-IP) and quantitative proteomics using Sequential Window Acquisition of all THeoretical mass spectra (SWATH-MS). Fifty-one proteins were enriched at least four-fold, p < 0.01, relative to cells transfected with an empty FLAG vector. A high proportion (24/51) of candidate CAPN5 interaction partners are associated with protein quality control, including components of the chaperonin, chaperone, and ubiquitin–proteasome systems. Additional candidate interactors include tubulins, kinases, phosphatases, G proteins, and mitochondrial proteins. CAPN5 interactions for 14 of the candidate proteins were confirmed by co-IP and immunoblotting. Of these 14 proteins, 11 exhibited in vitro calcium-induced proteolysis following co-IP with WT CAPN5-3×FLAG. Impaired calcium-induced proteolysis of co-IP proteins was observed for the pathogenic CAPN5 variants R243L and R289W. Further studies are needed to validate the association of candidate CAPN5 interactors with proteins and complexes suggested by the SWATH-MS and co-IP results, and the possible role of CAPN5 within such complexes. The possible involvement of CAPN5 in protein quality control is relevant to NIV, as defects in protein quality control have been implicated in inherited retinal disorders. Proteomic data are available via ProteomeXchange with identifier PXD068008. Full article
(This article belongs to the Special Issue Role of Calpains in Health and Diseases)
Show Figures

Figure 1

29 pages, 3045 KB  
Review
Plasmablasts as Translational Biomarkers in Autoimmune Diseases: From Cellular Dynamics to Clinical Decision-Making
by Muhammad Soyfoo and Julie Sarrand
Curr. Issues Mol. Biol. 2026, 48(1), 77; https://doi.org/10.3390/cimb48010077 - 12 Jan 2026
Viewed by 152
Abstract
B cells are key drivers of immune dysregulation across systemic autoimmune diseases. Among their progeny, plasmablasts occupy a uniquely revealing niche: short-lived, highly proliferative intermediates that mirror real-time B-cell activation. Their appearance in peripheral blood integrates antigenic stimulation, cytokine-driven differentiation, and aberrant germinal-center [...] Read more.
B cells are key drivers of immune dysregulation across systemic autoimmune diseases. Among their progeny, plasmablasts occupy a uniquely revealing niche: short-lived, highly proliferative intermediates that mirror real-time B-cell activation. Their appearance in peripheral blood integrates antigenic stimulation, cytokine-driven differentiation, and aberrant germinal-center dynamics, transforming them into sensitive indicators of ongoing immunological activity. This review synthesizes current knowledge on plasmablast biology and highlights disease-specific phenotypes across systemic lupus erythematosus (SLE), primary Sjögren disease (pSjD), IgG4-related disease (IgG4-RD), ANCA-associated vasculitis (AAV), and rheumatoid arthritis (RA). We incorporate molecular insights from single-cell technologies that have uncovered previously unrecognized plasmablast subsets, metabolic states, and interferon-related signatures with prognostic and mechanistic value. Beyond descriptive immunology, plasmablasts are emerging as dynamic biomarkers capable of informing real-time clinical decisions. One of the most robustly supported applications is the prognostic interpretation of plasmablast kinetics following B-cell-depleting therapies, where early reconstitution patterns consistently predict relapse across multiple autoimmune conditions. As clinical immunology shifts from static serological markers toward kinetic, cell-based monitoring, plasmablast quantification offers a path toward precision immune surveillance. Integrating plasmablast dynamics into routine care may ultimately allow clinicians to anticipate disease flares, time therapeutic reinforcements, and transition from reactive management to preventive intervention. Full article
Show Figures

Figure 1

19 pages, 1209 KB  
Review
Emerging Cell-Based Therapies for Systemic Sclerosis: From Stem Cells to CAR-T Cells
by Vitaly Chasov, Sabir Mukhametshin, Elvina Gilyazova, Damir Davletshin, Mariya Tikhomirova, Iuliia Topchu, Aygul Valiullina, Marcella Prete and Emil Bulatov
Curr. Issues Mol. Biol. 2026, 48(1), 76; https://doi.org/10.3390/cimb48010076 - 12 Jan 2026
Viewed by 138
Abstract
Systemic sclerosis (SSc) is a disease in which malfunctioning immune cells lead to the formation of autoantibodies that damage blood vessels and body tissues. Fibrosis then develops in the affected organs. Its complex pathogenesis involves multiple immune and stromal cell types, soluble mediators, [...] Read more.
Systemic sclerosis (SSc) is a disease in which malfunctioning immune cells lead to the formation of autoantibodies that damage blood vessels and body tissues. Fibrosis then develops in the affected organs. Its complex pathogenesis involves multiple immune and stromal cell types, soluble mediators, and dysregulated tissue repair, resulting in heterogeneous clinical manifestations and poor prognosis. Current disease-modifying therapies provide only modest benefits, often slowing but rarely reversing disease progression, and are associated with considerable adverse effects. These limitations have spurred the development of cell-based therapeutic strategies aimed at restoring immune tolerance and promoting tissue repair. In this review, we summarize recent advances in hematopoietic stem cell transplantation, mesenchymal stem cell therapy, and adoptive regulatory T cell transfer and highlight the emerging role of chimeric antigen receptor (CAR)-T cell therapy as a transformative approach for SSc. Collectively, these evolving strategies hold the potential to improve survival, achieve durable remissions, and significantly enhance quality of life for patients with SSc. Full article
(This article belongs to the Special Issue Molecular Basis of Autoimmune Diseases)
Show Figures

Figure 1

Back to TopTop