[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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
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 (7,138)

Search Parameters:
Keywords = autoimmunity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 2797 KiB  
Article
Incidence and Impact of Myocarditis in Genetic Cardiomyopathies: Inflammation as a Potential Therapeutic Target
by Yulia Lutokhina, Elena Zaklyazminskaya, Evgeniya Kogan, Andrei Nartov, Valeriia Nartova and Olga Blagova
Genes 2025, 16(1), 51; https://doi.org/10.3390/genes16010051 (registering DOI) - 4 Jan 2025
Viewed by 197
Abstract
Background: Myocardial disease is an important component of the wide field of cardiovascular disease. However, the phenomenon of multiple myocardial diseases in a single patient remains understudied. Aim: To investigate the prevalence and impact of myocarditis in patients with genetic cardiomyopathies and to [...] Read more.
Background: Myocardial disease is an important component of the wide field of cardiovascular disease. However, the phenomenon of multiple myocardial diseases in a single patient remains understudied. Aim: To investigate the prevalence and impact of myocarditis in patients with genetic cardiomyopathies and to evaluate the outcomes of myocarditis treatment in the context of cardiomyopathies. Methods: A total of 342 patients with primary cardiomyopathies were enrolled. The study cohort included 125 patients with left ventricular non-compaction (LVNC), 100 with primary myocardial hypertrophy syndrome, 70 with arrhythmogenic right ventricular cardiomyopathy (ARVC), 60 with dilated cardiomyopathy (DCM), and 30 with restrictive cardiomyopathy (RCM). The diagnosis of myocarditis was based on data from myocardial morphological examination or a non-invasive diagnostic algorithm consisting of an analysis of clinical presentation, anti-cardiac antibody (Ab) titres, and cardiac MRI. Results: The prevalence of myocarditis was 74.3% in ARVC, 56.7% in DCM, 54.4% in LVNC, 37.5% in RCM, and 30.9% in HCM. Myocarditis had a primary viral or secondary autoimmune nature and manifested with the onset or worsening of chronic heart failure (CHF) and arrhythmias. Treatment of myocarditis in cardiomyopathies has been shown to stabilise or improve patient condition and reduce the risk of adverse outcomes. Conclusions: In cardiomyopathies, the genetic basis and inflammation are components of a single continuum, which forms a complex phenotype. In genetic cardiomyopathies, myocarditis should be actively diagnosed and treated as it is an important therapeutic target. Full article
(This article belongs to the Section Genetic Diagnosis)
Show Figures

Figure 1

Figure 1
<p>Graphical representation depicting the structure of the patients included in the study, taking into account the presence of mixed phenotypes.</p>
Full article ">Figure 2
<p>Results of morphological study of myocardium in different cardiomyopathies. (<b>a</b>,<b>b</b>) Myocardial changes in HCM in the form of bizarre shape of branching cardiomyocytes, small focal cardiosclerosis with neoangiogenesis, and lymphohistiocytic infiltrates in sclerosis foci; (<b>c</b>,<b>d</b>) myocarditis in HCM with productive capillarites and development of interstitial sclerosis: myocardium is divided by fibrous septa of unequal thickness into lobules, uneven hypertrophy of nuclei, vessels with swollen endothelium, and perivascular accumulations of lymphoid elements are noted, more than 14 in the field of view at high magnification; (<b>e</b>,<b>f</b>) picture of lymphohistiocytic infiltration in ARVC, pronounced total fibrous-fatty replacement of myocardium of LV, the area of preserved myocardium in some areas does not exceed 25%; (<b>g</b>,<b>h</b>) lymphohistiocytic infiltrates perivascularly and in the interstitium (<b>g</b>) in a patient with DCM within laminopathy, fatty tissue replacement of dead cardiomyocytes (<b>h</b>); (<b>i</b>,<b>j</b>) SARS-CoV-2-induced myocarditis in a patient with RCM caused by pathogenic or likely pathogenic variants in <span class="html-italic">MyBPC3</span> and <span class="html-italic">LZTR1</span> genes: marked lymphohistiocytic infiltration, areas of lipomatosis, dystrophic changes in cardiomyocytes; (<b>k</b>,<b>l</b>) Ab to SARS-CoV-2 nucleocapsid (<b>k</b>) and spike antigen (<b>l</b>). (<b>a</b>–<b>c</b>,<b>e</b>–<b>j</b>)—haematoxylin and eosin staining; (<b>d</b>)—Van Gieson picrofuchsin staining; (<b>k</b>,<b>l</b>)—immunohistochemical study; (<b>a</b>,<b>f</b>)—low magnification; (<b>b</b>–<b>e</b>,<b>g</b>–<b>l</b>)—high magnification.</p>
Full article ">Figure 3
<p>Titres of anti-cardiac antibodies in different cardiomyopathies, depending on the presence (M+) or absence (M−) of myocarditis; * - <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Kaplan–Meier curves for different genetic cardiomyopathies, depending on the presence or absence of myocarditis. Red colour—patients with a combination of cardiomyopathy and myocarditis, blue colour—patients with isolated cardiomyopathies.</p>
Full article ">Figure 5
<p>Spectrum of genes with pathogenic or likely pathogenic variants in different cardiomyopathies, depending on the presence (M+) or absence (M−) of myocarditis.</p>
Full article ">Figure 6
<p>Frequency of myocarditis in different causes of myocardial hypertrophy syndrome.</p>
Full article ">Figure 7
<p>Frequency of superimposed myocarditis, depending on the type of cardiomyopathy.</p>
Full article ">
14 pages, 555 KiB  
Article
Functional and Cognitive Impairment in Patients with Relapsing–Remitting Multiple Sclerosis: Cognitive Tests and Plasma Neurofilament Light Chain Levels
by Elina Polunosika, Joel Simren, Arta Akmene, Nikita Klimovskis, Kaj Blennow, Daina Pastare, Henrik Zetterberg, Renars Erts and Guntis Karelis
Medicina 2025, 61(1), 70; https://doi.org/10.3390/medicina61010070 - 3 Jan 2025
Viewed by 394
Abstract
Background and Objectives: Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system. The disease can manifest and progress with both physical and cognitive symptoms, affecting the patient’s daily activities. The aim of our study was to [...] Read more.
Background and Objectives: Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system. The disease can manifest and progress with both physical and cognitive symptoms, affecting the patient’s daily activities. The aim of our study was to investigate the correlation between functional status, cognitive functions, and neurofilament light chain levels in plasma in MS patients. Materials and Methods: In a cross-sectional study, MS patients with a relapsing–remitting course (according to McDonald’s criteria, 2017) (n = 42) from Riga East University Hospital and a control group (n = 42) were included. In the MS group, the functional status was determined using the Expanded Disability Status Scale (EDSS), and neurofilament light chain levels in plasma (pNfL) were detected using single molecule array (Simoa) technology. The symbol digit modalities test (SDMT), brief visuospatial memory test—revised (BVMT-R), and the nine-hole peg test (9-HPT) were performed on the MS and control groups, dividing the groups by education level. Results: On the SDMT spreading speed, the MS group performed worse than the control group. The median score for the control group was 94.0, and for the MS group, it was 81.3. Slower performance on the SDMT also correlated with a higher EDSS in the MS group. Cognitive processing speed and memory were better in the control group and among individuals with higher education in both groups. For the BVMT-R, we found no difference between the two groups; both groups were able to learn the task equally well, but we found a weak correlation between age and learning in both groups, which could be related to the normal aging process. Execution reaction speed on the 9-HPT with the dominant hand was slower in the MS group (24.1 s) than in the control group (19.4 s). In the MS group, we observed a trend between SDMT performance and pNfL levels: higher pNfL levels were found in individuals who performed more slowly on the SDMT. Conclusions: Cognitive and fine motor dysfunction correlates with neurological impairment and plasma neurofilament light chain levels in MS patients. Full article
(This article belongs to the Section Neurology)
17 pages, 3803 KiB  
Article
Interstitial Lung Disease Associated with Anti-Ku Antibodies: A Case Series of 19 Patients
by Laure Petitgrand, Kaïs Ahmad, Delphine Gamondès, Rémi Diesler, Nicole Fabien, Laure Gallay, Romain Fort, Julie Traclet, François Lestelle, Roland Chapurlat, Cyrille B. Confavreux, Stéphane Durupt, Ségolène Turquier, Salim Aymeric Si-Mohamed, Frédéric Coutant and Vincent Cottin
J. Clin. Med. 2025, 14(1), 247; https://doi.org/10.3390/jcm14010247 - 3 Jan 2025
Viewed by 260
Abstract
Background: Antibodies against Ku have been described in patients with various connective tissue diseases. The objective of this study was to describe the clinical, functional, and imaging characteristics of interstitial lung disease in patients with anti-Ku antibodies. Methods: This single-center, retrospective observational [...] Read more.
Background: Antibodies against Ku have been described in patients with various connective tissue diseases. The objective of this study was to describe the clinical, functional, and imaging characteristics of interstitial lung disease in patients with anti-Ku antibodies. Methods: This single-center, retrospective observational study was conducted at a tertiary referral institution. Patients with positive anti-Ku antibodies and interstitial lung disease identified between 2007 and 2022 were included. Clinical, immunological, functional, and imaging data were systematically reviewed. Results: Nineteen patients (ten females) with a mean age of 59 ± 12.6 years were included. The most frequent associated diagnosis was systemic sclerosis (42%), followed by rheumatoid arthritis (26%), Sjögren syndrome, undifferentiated connective tissue disease, and overlap between systemic sclerosis and idiopathic inflammatory myopathy (scleromyositis). Imaging revealed frequent septal and intralobular reticulations and ground-glass opacities, with nonspecific interstitial pneumonia as the predominant pattern (53%). The mean forced vital capacity was 82% ± 26 of the predicted value, and the mean diffusing capacity for carbon monoxide was 55% ± 21. Over the first year of follow-up, the mean annual forced vital capacity decline was 140 mL/year (range: 0–1610 mL/year). The overall survival rate was 82% at 5 years and 67% at 10 years. Conclusions: Most patients with interstitial lung disease and anti-Ku antibodies presented with dyspnea, a mild-to-moderate restrictive ventilatory pattern, and reduced diffusing capacity for carbon monoxide. The CT pattern was heterogeneous but was consistent with nonspecific interstitial pneumonia in half of the patients. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the study. CT: computed tomographyl; ILD: interstitial lung disease.</p>
Full article ">Figure 2
<p>Characteristic speckled nuclear fluorescence pattern of anti-Ku antibodies observed on HEp-2 cells by indirect immunofluorescence. The white arrow highlights the nucleus of a cell in metaphase with fluorescence around the chromatin and negative mitoses.</p>
Full article ">Figure 3
<p>Computed tomography of a male patient with anti-Ku antibodies, demonstrating a pattern of fibrotic nonspecific interstitial pneumonia. (<b>A</b>) Axial view of lower part of the lungs, demonstrating diffuse distribution of ground-glass opacities, reticulation, bronchiectasis, and bronchiolectasis. (<b>B</b>) Sagittal view showing apicobasal distribution. (<b>C</b>) Coronal view, with minimal intensity projection of 13 mm. (<b>D</b>) Sagittal view, with minimal intensity projection of 13 mm.</p>
Full article ">Figure 4
<p>Computed tomography of a male patient with anti-Ku antibodies, demonstrating a pattern of fibrotic nonspecific interstitial pneumonia, apicobasal distribution of ground-glass opacities and reticulation with subpleural sparing, bronchiectasis, and bronchiolectasis, associated with paraseptal emphysema. (<b>A</b>) Upper part of the lungs, axial view. (<b>B</b>) Middle part of the lungs, axial view. (<b>C</b>) Lower part of the lungs, axial view. (<b>D</b>) Sagittal view, with a minimal intensity projection of 5 mm.</p>
Full article ">Figure 5
<p>Kaplan–Meier estimates of overall survival. The solid line represents the estimate of overall survival. The dotted lines represent the 95% confidence interval.</p>
Full article ">
13 pages, 1335 KiB  
Article
Clinical Profiles, Survival, and Lung Function Outcomes in ANCA-Associated Interstitial Lung Disease: An Observational Study
by Cristina Valero-Martínez, Claudia Valenzuela, Juan Pablo Baldivieso Achá, Elisa Martínez-Besteiro, Patricia Quiroga-Colina, Arantzazu Alfranca, Esther F. Vicente-Rabaneda, Susana Hernández Muñiz, Santos Castañeda and Rosario García-Vicuña
J. Clin. Med. 2025, 14(1), 229; https://doi.org/10.3390/jcm14010229 - 3 Jan 2025
Viewed by 222
Abstract
Background/Objectives: Anti-neutrophil cytoplasmic antibodies (ANCAs) have been found in interstitial lung disease (ILD) in recent years, although its impact on ILD prognosis is less known. To date, ANCAs are not included in the interstitial pneumonia with autoimmune features (IPAF) definition criteria. Therefore, [...] Read more.
Background/Objectives: Anti-neutrophil cytoplasmic antibodies (ANCAs) have been found in interstitial lung disease (ILD) in recent years, although its impact on ILD prognosis is less known. To date, ANCAs are not included in the interstitial pneumonia with autoimmune features (IPAF) definition criteria. Therefore, ANCA-ILD, in the absence of known ANCA-associated vasculitis (AAV), could be underdiagnosed. Our aim was to analyze the clinical profile and prognosis of ANCA-ILD patients. Methods: Patients diagnosed with ILD and positive ANCA were enrolled in a retrospective, monocentric cohort study. Lung function outcomes and mortality were assessed according to clinical, serological, radiological, and treatment characteristics. Survival was analyzed using Kaplan–Meier curves and Cox regression models. Results: A total of 23 patients were included, mostly women, with a median time from ILD diagnosis of 36 (24–68) months and a predominant anti-MPO pattern (56.5%). Nearly half of the patients had AAV, mostly microscopic polyangiitis (MPA). The presence of AAV was significantly associated with anti-MPO antibodies and an NSIP radiographic pattern. Overall, the fibrotic pattern (either UIP or fibrotic NSIP) was the most common (73.9%), mainly UIP (51.2%). However, it appeared less frequently in the AAV-ILD group. During follow-up, lung function impairment or radiological progression was observed in 65.2% of patients. Cumulative mortality incidence was high (43.4%), largely due to ILD itself (80%). A UIP pattern was associated with a higher and earlier mortality (HR 34.4 [1.36–132]), while the use of immunosuppressants showed a trend towards lower ILD-related death. Conclusions: In our cohort, ANCA-ILD patients mostly presented with fibrotic patterns, with AAV in almost half of the cases and a high and early mortality rate, which suggests the need to assess ANCA in all ILD patients. Full article
Show Figures

Figure 1

Figure 1
<p>Inspiratory axial HRCT images (<b>A</b>–<b>C</b>) in a 65-year-old woman diagnosed with isolated ANCA-ILD and positive C-ANCA. Pictures show reticulation (arrows), traction bronchiectasis (arrowhead), and ground glass opacities (asterisks), with no craniocaudal gradient. Bilateral mosaic pattern (geographic regions of decreased lung density) can also be detected (circles). Expiratory CT (<b>D</b>) confirms the presence of air trapping, and the mosaic pattern is highlighted compared to C. Findings suggest a non-UIP fibrotic interstitial lung disease.</p>
Full article ">Figure 2
<p>Axial (<b>A</b>) and coronal (<b>B</b>) high-resolution computed tomography images showing extensive fibrotic interstitial lung disease in a 70-year-old woman, diagnosed with isolated ANCA-ILD and anti-MPO positivity. Note the basilar predominance of honeycombing (arrows), traction bronchiectasis (arrowheads), and reticulation (asterisk); all findings are consistent with the usual interstitial pneumonia (UIP) pattern.</p>
Full article ">Figure 3
<p>Overall and stratified survival in the population with ILD and positive ANCA. (<b>A</b>) Kaplan–Meier survival curves for isolated ANCA-ILD or AAV_ILD based on the cumulative probability of survival. (<b>B</b>) Forest plot of the regression Cox analysis of the risk factors associated with premature mortality. Abbreviations: ANCA: anti-neutrophil cytoplasmic antibody; AAV: ANCA-associated vasculitis; Anti-MPO: anti-myeloperoxidase antibody; CI: confidence interval; HR: hazard ratio. ILD: interstitial lung disease.</p>
Full article ">
19 pages, 552 KiB  
Review
CAR-T Therapy Beyond B-Cell Hematological Malignancies
by Martina Canichella and Paolo de Fabritiis
Cells 2025, 14(1), 41; https://doi.org/10.3390/cells14010041 - 3 Jan 2025
Viewed by 323
Abstract
Despite the advances of CAR-T cells in certain hematological malignancies, mostly from B-cell derivations such as non-Hodgkin lymphomas, acute lymphoblastic leukemia and multiple myeloma, a significant portion of other hematological and non-hematological pathologies can benefit from this innovative treatment, as the results of [...] Read more.
Despite the advances of CAR-T cells in certain hematological malignancies, mostly from B-cell derivations such as non-Hodgkin lymphomas, acute lymphoblastic leukemia and multiple myeloma, a significant portion of other hematological and non-hematological pathologies can benefit from this innovative treatment, as the results of clinical studies are demonstrating. The clinical application of CAR-T in the setting of acute T-lymphoid leukemia, acute myeloid leukemia, solid tumors, autoimmune diseases and infections has encountered limitations that are different from those of hematological B-cell diseases. To overcome these restrictions, strategies based on different molecular engineering platforms have been devised and will be illustrated below. The aim of this manuscript is to provide an overview of the CAR-T application in pathologies other than those currently treated, highlighting both the limits and results obtained with these settings. Full article
Show Figures

Figure 1

Figure 1
<p>Different CAR-T clinical applications.</p>
Full article ">
34 pages, 3355 KiB  
Review
Inborn Errors of Immunity Presenting with Early-Onset Severe Atopy
by Nipat Chuleerarux, Nadia Makkoukdji, Travis Satnarine, Jessica Elise Kuhn, Tanawin Nopsopon, Peerada Valyasevi, Fernanda Bellodi Schmidt, Gary Kleiner and Melissa Gans
Medicina 2025, 61(1), 62; https://doi.org/10.3390/medicina61010062 - 2 Jan 2025
Viewed by 584
Abstract
Inborn errors of immunity (IEIs), also known as primary immunodeficiencies, are a group of genetic disorders affecting the development and function of the immune system. While IEIs traditionally present with recurrent infections, an increasing number of cases manifest with early-onset severe atopy, including [...] Read more.
Inborn errors of immunity (IEIs), also known as primary immunodeficiencies, are a group of genetic disorders affecting the development and function of the immune system. While IEIs traditionally present with recurrent infections, an increasing number of cases manifest with early-onset severe atopy, including atopic dermatitis, food allergies, asthma, and allergic rhinitis—features that are often overlooked. This can lead to delayed diagnosis and treatment, which is crucial for IEI patients due to the risk of severe infections. We conducted a literature search and reviewed all IEIs that can present with early-onset severe atopy. The hallmark features of these disorders often include early-onset, persistent, and severe atopic dermatitis, food allergies, and recurrent episodes of asthma, which may be refractory to treatments. Additionally, we discuss the importance of recognizing such severe atopy as a potential indicator of an underlying immune deficiency, particularly when accompanied by unusual infections, growth failure, or autoimmunity. This review aims to raise awareness of this association and emphasize the need for early diagnosis and genetic testing in patients with atypical or treatment-resistant allergic diseases, allowing for more timely diagnosis of underlying immunodeficiencies and appropriate treatments. Full article
(This article belongs to the Special Issue Pitfalls and Challenges in Allergy Management)
Show Figures

Figure 1

Figure 1
<p>The CBM complex controls T cell signaling by the mammalian target of rapamycin (mTOR), nuclear factor kappa B (NF-κB), and c-Jun N-terminal kinase activation (JNK). The T cell receptor leads to the transportation of glutamine, initiating the mTOR pathway.</p>
Full article ">Figure 2
<p>(<b>a</b>) The JAK/STAT pathways contain various factors, including <span class="html-italic">STAT5,</span> which leads to myeloid activation, or <span class="html-italic">STAT3</span>, which upregulates transcription of the gene <span class="html-italic">ZNF341</span>. (<b>b</b>) TGF-β signaling activates ERBIN/STAT3 complexes, inhibiting SMAD transport into the cell nucleus.</p>
Full article ">Figure 3
<p>NF-κB signaling pathway, involving either NEMO in the canonical pathway or <span class="html-italic">RelB</span> in the non-canonical pathway, is crucial for cell proliferation and immunity.</p>
Full article ">Figure 4
<p>The cytoskeletal pathway activated via a T cell receptor contributes to actin assembly, which is necessary for regulatory T cell functioning, intracellular interactions, and NF-κB signaling.</p>
Full article ">Figure 5
<p>Mast cell degranulation may be triggered easily in PLCγ2 mutations, leading to spontaneous calcium influx that results in degranulation by sub-physiologic temperatures of all hematopoietic cells, except T cells.</p>
Full article ">Figure 6
<p>Cytokine signaling gets activated by various interleukins, which are critical for immune and inflammation regulation.</p>
Full article ">Figure 7
<p>The skin barrier is maintained by several components, such as keratinocytes, intercellular adhesion proteins, and protease inhibitors, like <span class="html-italic">SPINK5</span>.</p>
Full article ">Figure 8
<p>Lymphocyte development involves many significant proteins, such as <span class="html-italic">RAG1</span>&amp;2, which are required for appropriate V(D)J recombination, in which mutations can lead to both B and T cell maturation failure.</p>
Full article ">Figure 9
<p>Regulatory T cell signaling requires the STAT5 pathway to encode the <span class="html-italic">FOXP3</span> gene, which is needed for appropriate T cell functioning and regulatory T cell differentiation.</p>
Full article ">Figure 10
<p>Mutations in the Phosphoacetylglucosamine Mutase 3 can lead to improper glycosylation, resulting in various conditions, such as hyper-IgE syndrome-like findings.</p>
Full article ">
13 pages, 1353 KiB  
Article
Mortality in Antinuclear Antibody-Positive Patients with and Without Rheumatologic Immune-Related Disorders: A Large-Scale Population-Based Study
by Uria Shani, Paula David, Ilana Balassiano Strosberg, Ohad Regev, Mohamad Yihia, Niv Ben-Shabat, Dennis McGonagle, Orly Weinstein, Howard Amital and Abdulla Watad
Medicina 2025, 61(1), 60; https://doi.org/10.3390/medicina61010060 - 2 Jan 2025
Viewed by 359
Abstract
Background & Objectives: To explore the potential association between positive ANA serology and all-cause mortality in a large cohort of patients, including those with and without rheumatological conditions and other immune-related diseases. Material and Methods: A retrospective cohort study analyzed all-cause [...] Read more.
Background & Objectives: To explore the potential association between positive ANA serology and all-cause mortality in a large cohort of patients, including those with and without rheumatological conditions and other immune-related diseases. Material and Methods: A retrospective cohort study analyzed all-cause mortality among 205,862 patients from Clalit Health Services (CHS), Israel’s largest health maintenance organization (HMO). We compared patients aged 18 and older with positive ANA serology (n = 102,931) to an equal number of ANA-negative controls (n = 102,931). Multivariable Cox regression models were used to assess hazard ratios (HR) for mortality, adjusting for demographic and clinical factors. Results: ANA positivity was strongly associated with increased mortality (adjusted HR [aHR] 4.62; 95% CI 4.5–4.7, p < 0.001). Significant predictors of mortality included male gender (39.2% vs. 24.4%, p < 0.001), older age at testing (72.4 ± 13.0 vs. 50.1 ± 17.3 years, p < 0.001), and Jewish ethnicity (89.6% vs. 83.2%, p < 0.001). Certain ANA patterns, such as mitochondrial (and dense fine speckled (DFS-AC2)), were highly predictive of mortality, with aHRs of 36.14 (95% CI 29.78–43.85) and 29.77 (95% CI 26.58–33.34), respectively. ANA-positive patients with comorbid rheumatological immune-related disorders (RIRDs) demonstrated a higher survival rate compared to those without such a condition (aHR 0.9, 95% CI 0.86–0.95, p < 0.001). This finding remained significant after adjusting for several parameters, including age. Conclusions: ANA positivity is associated with increased all-cause mortality, particularly in individuals without rheumatologic disorders, after adjusting for confounders such as age. This may indicate occult malignancies, cardiovascular pathology, or chronic inflammatory states, necessitating more vigilant surveillance Full article
(This article belongs to the Special Issue Autoimmune Diseases: Advances and Challenges)
Show Figures

Figure 1

Figure 1
<p>Survival analysis of patients with positive ANA compared to patients with negative ANA.</p>
Full article ">Figure 2
<p>Survival rates among different ANA patterns (<b>A</b>) and different ANA titers (<b>B</b>).</p>
Full article ">Figure 3
<p>Survival rates among patients with positive RIRD (Rheumatological immune-related disorder) status compared to asymptomatic patients. HR = Hazard Ratio, CI = Confidence Interval. Cox Regression adjusted to age, ethnicity, sex, and comorbidities.</p>
Full article ">
19 pages, 3453 KiB  
Article
Defining Mechanistic Links Between the Non-Coding Variant rs17673553 in CLEC16A and Lupus Susceptibility
by Harikrishna Reddy Rallabandi, Manish Kumar Singh, Loren L. Looger and Swapan K. Nath
Int. J. Mol. Sci. 2025, 26(1), 314; https://doi.org/10.3390/ijms26010314 - 1 Jan 2025
Viewed by 289
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by widespread inflammation and autoantibody production. Its development and progression involve genetic, epigenetic, and environmental factors. Although genome-wide association studies (GWAS) have repeatedly identified a susceptibility signal at 16p13, its fine-scale source and [...] Read more.
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by widespread inflammation and autoantibody production. Its development and progression involve genetic, epigenetic, and environmental factors. Although genome-wide association studies (GWAS) have repeatedly identified a susceptibility signal at 16p13, its fine-scale source and its functional and mechanistic role in SLE remain unclear. We used bioinformatics to prioritize likely functional variants and validated the top candidate through various experimental techniques, including clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing in B cells. To assess the functional impact of the proposed causal variant in C-type lectin domain family 16, member A (CLEC16A), we compared autophagy levels between wild-type (WT) and knock-out (KO) cells. Systematic bioinformatics analysis identified the highly conserved non-coding intronic variant rs17673553, with the risk allele apparently affecting enhancer function and regulating several target genes, including CLEC16A itself. Luciferase reporter assays followed by chromatin immunoprecipitation-quantitative polymerase chain reaction (ChIP-qPCR) validated this enhancer activity, demonstrating that the risk allele increases the binding of enhancer histone marks (H3K27ac and H3K4me1), the CTCF-binding factor, and key immune transcription factors (GATA3 and STAT3). Knock-down of GATA3 and STAT3 via siRNA led to a significant decrease in CLEC16A expression. These regulatory effects on the target gene were further confirmed using CRISPR-based genome editing and CRISPR-dCas9-based epigenetic activation/silencing. Functionally, WT cells exhibited higher levels of starvation-induced autophagy compared to KO cells, highlighting the role of CLEC16A and the rs17673553 locus in autophagy regulation. These findings suggest that the rs17673553 locus—particularly the risk allele—drives significant allele-specific chromatin modifications and binding of multiple transcription factors, thereby mechanistically regulating the expression of target autophagy-associated genes, including CLEC16A itself. This mechanism could potentially explain the association between rs17673553 and SLE, and could underlie the signal at 16p13. Full article
Show Figures

Figure 1

Figure 1
<p>Allele-specific activity of rs17673553. (<b>A</b>) Luciferase reporter assay results show allele-specific enhancer activity in non-immune (HEK293) and immune (Namalwa, Jurkat) cells. The risk allele (AA) showed significantly higher (Namalwa, <span class="html-italic">p</span> &lt; 0.0001; Jurkat, <span class="html-italic">p</span> &lt; 0.001; and HEK293, <span class="html-italic">p</span> &lt; 0.01) expression than the non-risk (GG) allele. (<b>B</b>) ChIP assay results showing allele-specific binding to DNA-binding proteins. Histone marks H3K27ac (<span class="html-italic">p</span> &lt; 0.0001) and H3K4me1 (<span class="html-italic">p</span> &lt; 0.0001), CTCF (<span class="html-italic">p</span> &lt; 0.0001), STAT3 (<span class="html-italic">p</span> &lt; 0.0001), and GATA3 (<span class="html-italic">p</span> &lt; 0.01) showed significantly higher binding to risk allele (AA) than non-risk allele (GG). Data represent mean ± SD from three biological replicates (n = 3). Statistical analysis was performed using GraphPad Prism (Version 10). (ns—not significant; *—<span class="html-italic">p</span> &lt; 0.05; **—<span class="html-italic">p</span> &lt; 0.005; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 2
<p>siRNA-based <span class="html-italic">GATA3</span> knockdown. To investigate the role of GATA3 at the rs17673553 locus, GATA3 was knocked down using siRNA in Namalwa cells. (<b>A</b>) A combined experiment of siRNA-based GATA3 knockdown and luciferase reporter assay results showing a significant decline in luciferase expression in both risk-AA (<span class="html-italic">p</span> &lt; 0.0001) and non-risk-GG (<span class="html-italic">p</span> &lt; 0.0001) compared to control. (<b>B</b>) qRT-PCR of luciferase, <span class="html-italic">GATA3</span>, <span class="html-italic">CLEC16A</span>, and <span class="html-italic">GAPDH</span>. siRNA transfection significantly reduced <span class="html-italic">GATA3</span> (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001, 0.0001), and luciferase (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), with no difference observed in negative control <span class="html-italic">GAPDH</span>. (<b>C</b>) qRT-PCR analysis of <span class="html-italic">GATA3</span> knockdown in Namalwa cells showed significant reduction in <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001) and <span class="html-italic">GATA3</span> (<span class="html-italic">p</span> &lt; 0.0001), and an increase in nearby genes <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001), with housekeeping gene <span class="html-italic">GAPDH</span> showing no difference. Data represent mean ± SD, values and statistical significance calculated using two-way ANOVA in GraphPad Prism (Version 10). (ns—not significant; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 2 Cont.
<p>siRNA-based <span class="html-italic">GATA3</span> knockdown. To investigate the role of GATA3 at the rs17673553 locus, GATA3 was knocked down using siRNA in Namalwa cells. (<b>A</b>) A combined experiment of siRNA-based GATA3 knockdown and luciferase reporter assay results showing a significant decline in luciferase expression in both risk-AA (<span class="html-italic">p</span> &lt; 0.0001) and non-risk-GG (<span class="html-italic">p</span> &lt; 0.0001) compared to control. (<b>B</b>) qRT-PCR of luciferase, <span class="html-italic">GATA3</span>, <span class="html-italic">CLEC16A</span>, and <span class="html-italic">GAPDH</span>. siRNA transfection significantly reduced <span class="html-italic">GATA3</span> (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001, 0.0001), and luciferase (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), with no difference observed in negative control <span class="html-italic">GAPDH</span>. (<b>C</b>) qRT-PCR analysis of <span class="html-italic">GATA3</span> knockdown in Namalwa cells showed significant reduction in <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001) and <span class="html-italic">GATA3</span> (<span class="html-italic">p</span> &lt; 0.0001), and an increase in nearby genes <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001), with housekeeping gene <span class="html-italic">GAPDH</span> showing no difference. Data represent mean ± SD, values and statistical significance calculated using two-way ANOVA in GraphPad Prism (Version 10). (ns—not significant; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 3
<p>siRNA-based STAT3-knockdown. To investigate the role of STAT3 at the rs17673553 locus, STAT3 was knocked down using siRNA in Namalwa cells. (<b>A</b>) A combined experiment of siRNA-based STAT3 knockdown and luciferase reporter assay results showing a significant decline in luciferase expression in both risk-AA (<span class="html-italic">p</span> &lt; 0.0001) and non-risk-GG (<span class="html-italic">p</span> &lt; 0.0001) compared to control. (<b>B</b>) qRT-PCR of luciferase, <span class="html-italic">GATA3, CLEC16A</span> and <span class="html-italic">GAPDH</span>. siRNA transfection significantly reduced <span class="html-italic">STAT3</span> (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001, 0.0001), and luciferase (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), with no difference observed in negative control <span class="html-italic">GAPDH</span>. (<b>C</b>) Comparative WT and STAT3-KD qRT-PCR results of <span class="html-italic">STAT3</span> knockdown in Namalwa cells showing strong reduction in the genes surrounding rs17673553 including <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001) along with <span class="html-italic">STAT3</span> (<span class="html-italic">p</span> &lt; 0.0001), with housekeeping gene <span class="html-italic">GAPDH</span> showing no difference. Data represent mean ± SD from at least three biological replicates (n = 3), values and statistical significance calculated using two-way ANOVA in GraphPad Prism (Version 10). (ns—not significant; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 3 Cont.
<p>siRNA-based STAT3-knockdown. To investigate the role of STAT3 at the rs17673553 locus, STAT3 was knocked down using siRNA in Namalwa cells. (<b>A</b>) A combined experiment of siRNA-based STAT3 knockdown and luciferase reporter assay results showing a significant decline in luciferase expression in both risk-AA (<span class="html-italic">p</span> &lt; 0.0001) and non-risk-GG (<span class="html-italic">p</span> &lt; 0.0001) compared to control. (<b>B</b>) qRT-PCR of luciferase, <span class="html-italic">GATA3, CLEC16A</span> and <span class="html-italic">GAPDH</span>. siRNA transfection significantly reduced <span class="html-italic">STAT3</span> (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001, 0.0001), and luciferase (<span class="html-italic">p</span> &lt; 0.0001, &lt;0.0001), with no difference observed in negative control <span class="html-italic">GAPDH</span>. (<b>C</b>) Comparative WT and STAT3-KD qRT-PCR results of <span class="html-italic">STAT3</span> knockdown in Namalwa cells showing strong reduction in the genes surrounding rs17673553 including <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001) along with <span class="html-italic">STAT3</span> (<span class="html-italic">p</span> &lt; 0.0001), with housekeeping gene <span class="html-italic">GAPDH</span> showing no difference. Data represent mean ± SD from at least three biological replicates (n = 3), values and statistical significance calculated using two-way ANOVA in GraphPad Prism (Version 10). (ns—not significant; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 4
<p>CRISPR-based epigenetic modification. CRISPR-dCas9 plasmids were co-transfected along with the guide RNA cloned vector. qRT-PCR results show that activation dCas9-vectors VPR significantly enhanced the expression of <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001); no notable impact was observed on <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> = 0.63). TET1 significantly increased the expression of <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001). Inhibitory dCas9-vectors MECP2 significantly suppressed <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001) and <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), with no observable effects on <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> = 0.58) and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> = 0.89). Similarly, DNMT3A suppressed <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001), CIITA (<span class="html-italic">p</span> &lt; 0.0001), and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001) expression, with no significant impact on <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> = 0.23). Data represent mean ± SD from three biological replicates (n = 3) and were analyzed using two-way ANOVA in GraphPad Prism (Version 10). (ns—not significant; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 5
<p>CRISPR knockout of <span class="html-italic">CLEC16A.</span> (<b>A</b>) A 183bp sequence surrounding rs17673553 was knocked out using CRISPR-Cas9. Genomic coordinates of CLEC16A (upper panel) and the intronic SNP. Comparative results (lower panel) showing the 2 sgRNAs and the CRSIPR-knockout region from CLEC16A-KO cells and SNP-containing sequence from wild-type cells. (<b>B</b>) qRT-PCR results showing the effect of the SNP region knockout on target gene <span class="html-italic">CLEC16A</span> and other nearby genes. Significantly lower expression of target gene <span class="html-italic">CLEC16A</span> (<span class="html-italic">p</span> &lt; 0.0001) and adjacent genes <span class="html-italic">DEXI</span> (<span class="html-italic">p</span> &lt; 0.0001), <span class="html-italic">CIITA</span> (<span class="html-italic">p</span> &lt; 0.001) and <span class="html-italic">RMI2</span> (<span class="html-italic">p</span> &lt; 0.0001) was seen in KO cells than WT cells. A neighboring control gene <span class="html-italic">KLF2</span> (<span class="html-italic">p</span> = 0.999) had no significant impact on its expression. (<b>C</b>) Western blot analysis showing the differential expression of <span class="html-italic">CLEC16A</span> in KO (lane1) and WT (lane2) cells, as well as in non-risk (18566) and risk (18624) LCL cells. (<b>D</b>) Densitometry analysis of Western blotting showing CLEC16A protein expression difference (fold) between WT-KO and risk/non-risk samples relative to a GAPDH control. Data represent mean ± SD from at least three biological replicates (n = 3). (ns—not significant; **—<span class="html-italic">p</span> &lt; 0.005; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">Figure 6
<p>(<b>A</b>). Starvation-induced autophagy. qRT-PCR assessment of EBSS-starved CLEC16A-WT and CLEC16-KO Namalwa cells showing differential expression of autophagy-related genes. Both WT and <span class="html-italic">CLEC16A-KO</span> cells were starved and analyzed for the samples at 0 h (basal), 2 h, 4 h, and 6 h regular intervals. Pairwise comparison between WT and KO showing <span class="html-italic">CLEC16A</span> expression is reduced across the samples at 0 h being slightly low (<span class="html-italic">p</span> = 0.003), significant reduction at 2 h (<span class="html-italic">p</span> &lt; 0.001), 4 h (<span class="html-italic">p</span> &lt; 0.001), and at 6 h it is greatly reduced in both samples (<span class="html-italic">p</span> = 0.012). Autophagy-related genes <span class="html-italic">LC3A, LC3B, ULK1</span>, and <span class="html-italic">P62</span> have enhanced expression in KO cells compared to WT cells, at 0 h showing slight increase in <span class="html-italic">LC3A</span> (<span class="html-italic">p</span> = 0.003) and <span class="html-italic">LC3B</span> (<span class="html-italic">p</span> = 0.005) but with <span class="html-italic">ULK1</span> (<span class="html-italic">p</span> = 0.002) and <span class="html-italic">P62</span> (<span class="html-italic">p</span> = 0.006) being reduced. At 2 h, they showed significant enhanced expression in <span class="html-italic">LC3A</span> (<span class="html-italic">p</span> = 0.001), <span class="html-italic">ULK1</span> (<span class="html-italic">p</span> = 0.009) but no significant change was observed in <span class="html-italic">LC3B</span> (<span class="html-italic">p</span> = 0.066) and <span class="html-italic">P62</span> (<span class="html-italic">p</span> = 0.56). At 4 h the trend was also similarly followed with <span class="html-italic">LC3A</span> (<span class="html-italic">p</span> = 0.010), <span class="html-italic">LC3B</span> (<span class="html-italic">p</span> = 0.002), and <span class="html-italic">ULK1</span> (<span class="html-italic">p</span> = 0.009) but <span class="html-italic">P62</span> (<span class="html-italic">p</span> = 0.233) showing no significant difference. At 6 h, all autophagy genes showed greater enhanced expression in KO cells <span class="html-italic">LC3A</span> (<span class="html-italic">p</span> &lt; 0.001), <span class="html-italic">LC3B</span> (<span class="html-italic">p</span> &lt; 0.001), and <span class="html-italic">ULK1</span> (<span class="html-italic">p</span> &lt; 0.001), but <span class="html-italic">P62 (p</span> &lt; 0.001) significantly reduced. The autophagy-regulating gene <span class="html-italic">mTORC1</span> followed a similar trend to <span class="html-italic">CLEC16A</span> and showed lower expression at 0 h (<span class="html-italic">p</span> &lt; 0.001), 2 h (<span class="html-italic">p</span> &lt; 0.001), and 4 h (<span class="html-italic">p</span> &lt; 0.001), and both WT and KO cells showed greater down trends, but no significant difference was observed (<span class="html-italic">p</span> = 0.272). All the measurements were repeated at least three times (n = 3), calculated mean ± SD, and significance was calculated for pairwise comparisons using GraphPad Prism (Version 10) and multiple <span class="html-italic">t</span>-test per row method. (<b>B</b>) Starvation-induced autophagy in CLEC16A knockdown (KD) cells. qRT-PCR assessment of EBSS-starved CLEC16A-KD Namalwa cells showing the differential expression of autophagy-related genes. Comparison of EBSS-starved CLEC16A-KD cells with WT cells at a regular intervals of 0 h (basal), 2 h, 4 h, and 6 h denoting a significant reduction in autophagy-related genes. In KD cells <span class="html-italic">CLEC16A</span> gene expression (<span class="html-italic">p</span> &lt; 0.001) and <span class="html-italic">mTORC1</span> (<span class="html-italic">p</span> &lt; 0.001) were reduced but did not impact the other autophagy markers LC3B (<span class="html-italic">p</span> = 0.552), P62 (<span class="html-italic">p</span> = 0.191), and <span class="html-italic">ULK1</span> (<span class="html-italic">p</span> = 0.866) and were compared to wild type WT. Starved KD cells showed significantly enhanced expression of <span class="html-italic">LC3B</span> at 2 h, 4 h, and 6 h (<span class="html-italic">p</span> &lt; 0.001,0.001, 0.001), and similarly <span class="html-italic">ULK1</span> increased at 2 h, 4 h, and 6 h (<span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.01), but <span class="html-italic">P62</span> reduced at 2 h, 4 h, and 6 h (<span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">p</span> &lt; 0.001). <span class="html-italic">CLEC16A</span> showed significant down-trend at 2 h, 4 h, and 6 h (<span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.001), and <span class="html-italic">mTORC1</span> also significantly reduced at 2 h, 4 h, and 6 h (<span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.001). All the measurements were replicated (n = 3) and calculated using multiple t-test method on GraphPad Prism (Version 10). (ns—not significant; *—<span class="html-italic">p</span> &lt; 0.05; **—<span class="html-italic">p</span> &lt; 0.005; ***—<span class="html-italic">p</span> &lt; 0.001.)</p>
Full article ">
16 pages, 2656 KiB  
Article
Innovative Lipid Nanoparticles Co-Delivering Hydroxychloroquine and siRNA for Enhanced Rheumatoid Arthritis Therapy
by Yanru Feng, Xintong Pan, Ziqian Li, Yue Li, Ya’nan Sun, Shaokun Yang, Chaoxing He, Yunjie Dang, Lu Huang and Bai Xiang
Pharmaceutics 2025, 17(1), 45; https://doi.org/10.3390/pharmaceutics17010045 - 1 Jan 2025
Viewed by 313
Abstract
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (si [...] Read more.
Background: Rheumatoid arthritis (RA) is a debilitating autoimmune disorder characterized by chronic inflammation and joint damage. Despite advancements in treatment, complete remission remains elusive. Methods: In this study, we introduce a novel lipid nanoparticle formulation co-delivering hydroxychloroquine (HCQ) and siRNA targeting TNF-α (siTNF-α) using microfluidic technology, marking the first use of such a combination for RA therapy. Results: In LPS-stimulated RAW 264.7 cells, the nanoparticles effectively reduced inflammatory markers. When administered via an intra-articular injection in a rat model, they significantly decreased joint inflammation and demonstrated good biological safety. Conclusions: This pioneering approach highlights the potential of lipid nanoparticles as a dual-delivery platform for enhanced RA treatment through targeted intra-articular administration. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Characterization of LNP-si<span class="html-italic">TNF-α</span> and LNP-si<span class="html-italic">TNF-α</span>-HCQ. (<b>A</b>) Preparation process of LNP-si<span class="html-italic">TNF-α</span>-HCQ. Morphology of LNP-si<span class="html-italic">TNF-α</span> (<b>B</b>) and LNP-si<span class="html-italic">TNF-α</span>-HCQ (<b>C</b>) characterized by cryo-electron microscopy. (<b>D</b>) The high-performance liquid chromatogram of HCQ. (<b>E</b>) The serum stability of free si<span class="html-italic">N.C.</span>, LNP-si<span class="html-italic">N.C.</span>, and LNP-si<span class="html-italic">N.C.</span>-HCQ. (<b>F</b>) The particle size and PDI stability of LNP-si<span class="html-italic">TNF-α</span> and LNP-si<span class="html-italic">TNF-α</span>-HCQ.</p>
Full article ">Figure 2
<p>Cytotoxicity in RAW 264.7 cells of LNP-si<span class="html-italic">N.C.</span> (<b>A</b>), LNP-si<span class="html-italic">TNF-α</span> (<b>B</b>), LNP-si<span class="html-italic">N.C.</span>-HCQ (<b>C</b>), and LNP-si<span class="html-italic">TNF-α</span>-HCQ (<b>D</b>) prepared with different concentrations of siRNA; ns: no significance.</p>
Full article ">Figure 3
<p>The cellular uptake of free si<span class="html-italic">N.C.</span>-Cy5, LNP-si<span class="html-italic">N.C.</span>-Cy5, and LNP-si<span class="html-italic">N.C.</span>-Cy5-HCQ by RAW 264.7 cells was studied using confocal laser microscopy (<b>A</b>) and flow cytometry (<b>B</b>). (<b>C</b>) The in vitro anti-inflammatory effect of free si<span class="html-italic">TNF-α</span>, Free HCQ, LNP-si<span class="html-italic">TNF-α</span>, LNP-si<span class="html-italic">N.C.</span>-HCQ, and LNP-si<span class="html-italic">TNF-α</span>-HCQ on RAW 264.7 cells. Scale bar: 10 μm (<b>A</b>); ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns: no significance.</p>
Full article ">Figure 4
<p>Therapeutic efficacy of LNP-si<span class="html-italic">TNF-α</span>-HCQ on adjuvant-induced rheumatoid arthritis rat model. (<b>A</b>) Timeline of in vivo study. (<b>B</b>) Biodistribution of LNP-si<span class="html-italic">TNF-α</span>-HCQ in rheumatoid arthritis rat articular cavity. Arthritis score (<b>C</b>), paw volume (<b>D</b>), and TNF-α content (<b>F</b>) in plasma of AIA rats were assessed in different groups. (<b>E</b>) The picture of the rat’s hind paw in different groups before sacrifice. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Safety evaluation of AIA rats treated with different administration groups. (<b>A</b>) H&amp;E staining of various organs. (<b>B</b>) The body weight changes in rats. (<b>C</b>–<b>F</b>) The number of WBCs, RBCs, PLTs, and Lymphs in the blood after rats were sacrificed. (<b>G</b>–<b>J</b>) The concentration of AST, ALT, ALP, and CREA in the blood. Scale bar: 100 μm; ns: no significance.</p>
Full article ">
15 pages, 933 KiB  
Article
The Influence of Concurrent Autoimmune Thyroiditis on the Cardiometabolic Consequences of Cabergoline in Postmenopausal Women
by Robert Krysiak, Marcin Basiak, Witold Szkróbka and Bogusław Okopień
Metabolites 2025, 15(1), 9; https://doi.org/10.3390/metabo15010009 - 1 Jan 2025
Viewed by 401
Abstract
Background: Untreated hyperprolactinemia and autoimmune thyroiditis (Hashimoto’s disease) seem to increase cardiometabolic risk. The cardiometabolic effects of cabergoline were less significant in young women with concurrent euthyroid Hashimoto’s illness. This study sought to investigate if the detrimental effects of this condition on cabergoline [...] Read more.
Background: Untreated hyperprolactinemia and autoimmune thyroiditis (Hashimoto’s disease) seem to increase cardiometabolic risk. The cardiometabolic effects of cabergoline were less significant in young women with concurrent euthyroid Hashimoto’s illness. This study sought to investigate if the detrimental effects of this condition on cabergoline efficacy are also evident in postmenopausal women. Methods: The study comprised 50 postmenopausal women exhibiting increased prolactin levels, with half qualifying for euthyroid Hashimoto’s illness. The subjects with thyroid autoimmunity were matched with those without thyroid disease according to age, body mass index, and prolactin levels. In addition to prolactin, we assessed thyroid-stimulating hormone (TSH), thyroid antibodies, and glucose homeostasis markers: fasting glucose, the homeostatic model assessment 1 of insulin resistance ratio (HOMA1-IR), and glycated hemoglobin (HbA1c). Furthermore, we assessed plasma lipids, plasma uric acid levels, high-sensitivity C-reactive protein (hsCRP), fibrinogen, homocysteine, and the urine albumin-to-creatinine ratio (UACR). The decadal cardiovascular risk was assessed with the Framingham Risk Score (FRS). Results: Before therapy, disparities existed among groups in HOMA1-IR, HDL cholesterol, antibody titers, uric acid, hsCRP, fibrinogen, homocysteine, UACR, and FRS. After six months of treatment, cabergoline successfully corrected prolactin levels (both total and monomeric) in women without thyroid disorders. This normalization correlated with decreases in HOMA1-IR, triglycerides, uric acid, hsCRP, fibrinogen, homocysteine, UACR, and FRS, as well as an elevation in HDL cholesterol. In women diagnosed with Hashimoto’s disease, cabergoline’s effects were limited to a reduction in prolactin levels, HOMA1-IR, and UACR, as well as an elevation in HDL cholesterol, with these alterations being less pronounced compared to women without thyroid illness. Conclusions: The cardiometabolic benefits of cabergoline were associated with the degree of prolactin concentration reduction. In women diagnosed with Hashimoto’s disease, connections were noted between baseline levels and treatment-induced alterations in hsCRP. These data indicate that concurrent euthyroid autoimmune thyroiditis mitigates the cardiometabolic consequences of cabergoline. Full article
Show Figures

Figure 1

Figure 1
<p>Flow the patients through the study.</p>
Full article ">Figure 2
<p>Percentage changes from baseline in the investigated variables during cabergoline treatment. Group 1: women with prolactin excess and AT. Group 2: women with prolactin excess but without thyroid pathology. The <a href="#app1-metabolites-15-00009" class="html-app">Supplementary Files (Table S1)</a> present the full data as the mean and standard deviation. Abbreviations: AT—autoimmune thyroiditis; FRS—Framingham Risk Score; HbA1c—glycated hemoglobin; HDL—high-density lipoprotein; HOMA1-IR—the homeostatic model assessment 1 of insulin resistance ratio; hsCRP—high-sensitivity C-reactive protein; LDL—low-density lipoprotein; TgAbs—thyroglobulin antibodies; TPOAbs—thyroid peroxidase antibodies; TSH—thyroid-stimulating hormone; UACR—urinary albumin-to-creatinine ratio.</p>
Full article ">
21 pages, 1751 KiB  
Review
The Role of Oral Biomarkers in the Assessment of Noncommunicable Diseases
by Gustavo Sáenz-Ravello, Marcela Hernández, Mauricio Baeza and Patricia Hernández-Ríos
Diagnostics 2025, 15(1), 78; https://doi.org/10.3390/diagnostics15010078 - 31 Dec 2024
Viewed by 243
Abstract
Background/Objectives: Oral biomarkers have gained attention as non-invasive tools for assessing systemic diseases due to their potential to reflect physiological and pathological conditions. This review aims to explore the role of oral biomarkers in diagnosing and monitoring systemic diseases, emphasizing their diagnostic [...] Read more.
Background/Objectives: Oral biomarkers have gained attention as non-invasive tools for assessing systemic diseases due to their potential to reflect physiological and pathological conditions. This review aims to explore the role of oral biomarkers in diagnosing and monitoring systemic diseases, emphasizing their diagnostic relevance and predictive capabilities in clinical practice. Methods: This narrative review synthesizes the current literature on biochemical, immunological, genetic, and microbiological oral biomarkers, with a focus on their sources, types, and clinical applications. Key studies were analyzed to identify associations between oral biomarkers and systemic diseases such as cardiovascular diseases, type 2 diabetes mellitus, autoimmune disorders, and cancers. Results: Oral fluids, including saliva and gingival crevicular fluid, contain diverse biomarkers such as matrix metalloproteinases, cytokines, and genetic indicators. These markers have demonstrated potential in diagnosing and monitoring systemic conditions. Among others, elevated levels of salivary glucose and inflammatory cytokines correlate with diabetes progression, while vascular endothelial growth factor (VEGF) and salivary C-reactive protein might be applicable as indicators for periodontal disease and cardiovascular risk. Additionally, salivary biomarkers like amyloid-beta and tau are promising in detecting neurodegenerative disorders. Conclusions: Oral biomarkers might represent a transformative and point-of-care approach to the early management of systemic diseases; however, challenges in measurement variability, standardization, and validation remain. Full article
(This article belongs to the Special Issue Diagnostics and Management in Oral and Maxillofacial Medicine)
Show Figures

Figure 1

Figure 1
<p>Summary of main oral biomarkers synthesized in this review. Source: BioRender.</p>
Full article ">Figure 2
<p>Diagram showing relationships between oral biomarkers, periodontitis, and systemic diseases. Source: Self-elaboration.</p>
Full article ">Figure 3
<p>Percentage of patents of oral biomarkers for assessment of noncommunicable diseases by country (or organization). Source: data from Google Patents after searching for “oral biomarkers AND noncommunicable diseases” on 26 December 2024 (10,278 results). Source: Self-elaboration.</p>
Full article ">Figure 4
<p>The evolution of the number of patents of oral biomarkers for the assessment of noncommunicable diseases by year. Source: data from Google Patents after searching for “oral biomarkers AND noncommunicable diseases” on 26 December 2024 (10,278 results). Source: Self-elaboration.</p>
Full article ">
32 pages, 1569 KiB  
Review
Autoimmune Thyroid Disease and Pregnancy: The Interaction Between Genetics, Epigenetics and Environmental Factors
by Tatjana Bogović Crnčić, Božena Ćurko-Cofek, Lara Batičić, Neva Girotto, Maja Ilić Tomaš, Antea Kršek, Ines Krištofić, Tea Štimac, Ivona Perić, Vlatka Sotošek and Sanja Klobučar
J. Clin. Med. 2025, 14(1), 190; https://doi.org/10.3390/jcm14010190 - 31 Dec 2024
Viewed by 284
Abstract
Autoimmune thyroid disease (AITD) is the leading cause of thyroid dysfunction globally, characterized primarily by two distinct clinical manifestations: Hashimoto’s thyroiditis (HT) and Graves’ disease (GD). The prevalence of AITD is approximately twice as high in women compared to men, with a particularly [...] Read more.
Autoimmune thyroid disease (AITD) is the leading cause of thyroid dysfunction globally, characterized primarily by two distinct clinical manifestations: Hashimoto’s thyroiditis (HT) and Graves’ disease (GD). The prevalence of AITD is approximately twice as high in women compared to men, with a particularly pronounced risk during the reproductive years. Pregnancy exerts profound effects on thyroid physiology and immune regulation due to hormonal fluctuations and immune adaptations aimed at fostering maternal–fetal tolerance, potentially triggering or exacerbating AITD. The impact of AITD on pregnancy outcomes is multifaceted. Both HT and GD have been associated with adverse obstetric and neonatal outcomes, including miscarriage, preterm delivery, preeclampsia and fetal growth restriction. Inadequately managed AITD can also affect fetal neurodevelopment due to disrupted maternal thyroid hormone availability during critical periods of brain maturation. This review explores the complex interplay between the genetic, epigenetic and environmental factors that drive AITD during pregnancy, highlighting their roles in disease development and impacts on pregnancy outcomes. Gaining a deeper understanding of these mechanisms is crucial for improving diagnostic tools, treatment options and preventive measures to enhance the health and well-being of both the mother and the newborn. Full article
(This article belongs to the Special Issue Thyroid Disease: Updates from Diagnosis to Treatment)
Show Figures

Figure 1

Figure 1
<p>Assessment of thyroid function during pregnancy. The figure shows a schematic representation of thyroid function tests during pregnancy according to the American Thyroid Association (2017) [<a href="#B58-jcm-14-00190" class="html-bibr">58</a>]. Overt autoimmune hyperthyroidism is characterized by a thyroid stimulating hormone (TSH) level of less than 0.1 mU/L and elevated thyroid hormones and thyroid receptor antibodies (TRAbs), indicating the need for treatment. Transient gestational thyrotoxicosis is suspected when normal thyroid hormone levels are observed while TSH is suppressed and TRAb levels are in the reference range. If TRAb levels are elevated, subclinical autoimmune hyperthyroidism should be considered. In both cases, a follow-up examination is required. (Abbreviations: TSH—thyroid stimulating hormone, T4—thyroxine, FT4—free thyroxine, T3—triiodothyronine, FT3—free triiodothyronine, TRAbs—thyroid receptor antibodies, TPOAb—thyroid peroxidase antibody, and N—normal, ↑—elevated, ↓—decreased).</p>
Full article ">Figure 2
<p>Key factors influencing thyroid function. Figure illustrates the most critical physiological, environmental, genetic and lifestyle factors that impact thyroid function.</p>
Full article ">Figure 3
<p>The complex network of factors that influence thyroid function and pregnancy outcome. Both thyroid function and a successful pregnancy are influenced by genetic and epigenetic predispositions as well as environmental factors that, when out of balance, can lead to AITD and an adverse pregnancy outcome. Immunomodulation during pregnancy, which is necessary for a successful pregnancy outcome, also affects thyroid function, especially in the presence of AITD. On the other hand, autoimmunity of the thyroid gland can affect the normal immunological changes and the outcome of pregnancy.</p>
Full article ">
14 pages, 980 KiB  
Article
Gluten-Free Diet Adherence Evaluation in Adults with Long-Standing Celiac Disease
by Marek K. Kowalski, Danuta Domżał-Magrowska, Piotr Szcześniak, Magdalena Bulska, Daria Orszulak-Michalak and Ewa Małecka-Wojciesko
Foods 2025, 14(1), 76; https://doi.org/10.3390/foods14010076 - 31 Dec 2024
Viewed by 375
Abstract
Background: Celiac disease (CD) is an autoimmune disease that results from the interaction of genetic, immune, and environmental factors. According to the 2020 European Society for Pediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) guidelines, an elimination diet (i.e., excluding products that may contain gluten) [...] Read more.
Background: Celiac disease (CD) is an autoimmune disease that results from the interaction of genetic, immune, and environmental factors. According to the 2020 European Society for Pediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) guidelines, an elimination diet (i.e., excluding products that may contain gluten) is the basic method of treating celiac disease. Following a gluten-free diet is extremely problematic, and patients often make unconscious deviations from the diet. According to the current Oslo definitions for celiac disease, depending on the clinical picture and adequate tests, several forms of celiac disease have been identified: typical, atypical, asymptomatic, potential, and refractory. Objective: The aim of the study was to assess the frequency of conscious diet mistakes and unconscious deviations from a gluten-free diet in a group of patients with long-standing celiac disease and their impact on the frequency of typical and atypical symptoms. Methods: The study included 57 people diagnosed with celiac disease between 1980 and 2010. After verifying the history of the disease according to the ESPGHAN guidelines from 2020, we excluded 19 patients who had Marsh grade 1 at the time of diagnosis or those without HLA DQ2 or DQ8 haplotypes detected. After verification, the study included 38 patients, 30 women and 8 men, with a verified diagnosis of typical celiac disease. The effectiveness of the gluten-free diet was assessed in all participants. Blood was collected to determine IgA anti-tissue transglutaminase II antibodies (anti-tTG) and IgG antibodies against deamidated gliadin peptides by ELISA. All survey participants provided data concerning current gastrointestinal and systemic symptoms, bowel habits, comorbidities, dietary habits, physical activity, and socioeconomic conditions. Results: A total of 25 patients (65.78%) declared strict adherence to the gluten-free diet. However, in this group, seven (18.4%) patients had significantly increased levels of anti-tTG antibodies (mean 82.3 RU/mL ± 78.9 SD at N < 20 RU/mL). Among the patients who consciously made dietary mistakes, six (46.2%) demonstrated increased levels of anti-tTG antibodies. The analysis did not reveal any difference between the frequency of intestinal and extraintestinal symptoms in patients making dietary mistakes and following the gluten-free diet. Conclusions: More than half of celiac patients unconsciously or consciously make dietary mistakes, which indicates an urgent need to increase their general knowledge of CD and the appropriate diet. Regardless of whether the gluten-free diet is followed, both typical and atypical symptoms of the disease have been observed among celiac patients. Full article
(This article belongs to the Special Issue Gluten-Free Food and Celiac Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Declared adherence to the gluten-free diet in the group.</p>
Full article ">Figure 2
<p>Evaluation of the frequency of elevated levels of anti-tissue transglutaminase antibodies (&gt;20 UI/mL).</p>
Full article ">Figure 3
<p>Evaluation of the frequency of intestinal symptoms in patients with celiac disease depending on gluten-free diet adherence.</p>
Full article ">Figure 4
<p>Evaluation of the frequency of extraintestinal symptoms in celiac patients depending on gluten-free diet adherence.</p>
Full article ">
9 pages, 1091 KiB  
Article
Clinical Presentations of Celiac Disease: Experience of a Single Italian Center
by Chiara Maria Trovato, Francesca Ferretti, Anna Pia Delli Bovi, Giovanna Elefante, Monica Ancinelli, Giulia Bolasco, Teresa Capriati, Sabrina Cardile, Daniela Knafelz, Fiammetta Bracci, Arianna Alterio, Monica Malamisura, Salvatore Grosso, Paola De Angelis and Antonella Diamanti
Nutrients 2025, 17(1), 129; https://doi.org/10.3390/nu17010129 - 31 Dec 2024
Viewed by 292
Abstract
Background/Objectives: In Italy, the prevalence of celiac disease (CeD) among children exceeds 1.5% and has steadily increased with a linear trend over the past 25 years. The clinical presentation is heterogenous and a change in onset symptoms has been described in recent years. [...] Read more.
Background/Objectives: In Italy, the prevalence of celiac disease (CeD) among children exceeds 1.5% and has steadily increased with a linear trend over the past 25 years. The clinical presentation is heterogenous and a change in onset symptoms has been described in recent years. The aim of the study is to describe the pattern of clinical presentation of CeD during the last 12 years in a single Italian center. Methods: We retrospectively enrolled all children diagnosed with CeD at Bambino Gesù Children Hospital, Rome between 1 March 2011 and 22 June 2023. To investigate the changes in pattern of clinical presentation, we divided the patient population into three groups of approximately 4 years each (respectively: 49, 48 and 48 months). Patients who previously received a CeD diagnosis in other centers were excluded. Results: Overall, 4478 patients were diagnosed with CeD at our center. 1082 were excluded, leaving 3396 available for analysis. We divided the study cohort into three groups: group 1 (n = 909), group 2 (n = 1103), and group 3 (n = 1384). Diagnoses of CeD increased by 17.5%. The trend of the non-classic form shows a significative increase (p = 0.000064), showing a high prevalence of bloating and abdominal pain and a significant reduction in celiac crisis (p < 0.0001). Conclusions: Annual diagnoses of CeD increased during the study period, and the clinical presentation has changed in recent years, showing an increase in the non-classic form and a reduction in more severe forms of celiac crisis. Full article
(This article belongs to the Special Issue Nutrition and Immunobiology of Celiac Disease)
Show Figures

Figure 1

Figure 1
<p>Flowchart of study population.</p>
Full article ">Figure 2
<p>Distribution of age and sex at diagnosis in the three groups.</p>
Full article ">Figure 3
<p>Classic, non-classic and asymptomatic forms variation during the time. Group 1, <span class="html-italic">n</span>= 909, diagnosed between 1 March 2011 and 30 April 2015; group 2, <span class="html-italic">n</span>= 1103, diagnosed between 1 May 2015 and 30 May 2019; group 3, <span class="html-italic">n</span>= 1384, diagnosed between 1 June 2019 and 22 June 2023.</p>
Full article ">
13 pages, 541 KiB  
Perspective
Celiac Disease, Gluten-Free Diet, and Eating Disorders: From Bench to Bedside
by Yaohui Wei, Yating Wang, Yonggui Yuan and Jue Chen
Foods 2025, 14(1), 74; https://doi.org/10.3390/foods14010074 - 31 Dec 2024
Viewed by 383
Abstract
Celiac disease (CD) and eating disorders (EDs) are complex chronic conditions in adolescents, sharing symptoms such as weight change, malnutrition, and gastrointestinal symptoms. CD, an autoimmune disorder triggered by gluten ingestion, is managed through a strict gluten-free diet that can unintentionally foster disordered [...] Read more.
Celiac disease (CD) and eating disorders (EDs) are complex chronic conditions in adolescents, sharing symptoms such as weight change, malnutrition, and gastrointestinal symptoms. CD, an autoimmune disorder triggered by gluten ingestion, is managed through a strict gluten-free diet that can unintentionally foster disordered eating behaviors due to dietary restrictions. Conversely, EDs may mask and complicate CD symptoms, leading to diagnostic delays and treatment challenges. Evidence reveals an increased risk of EDs in CD individuals and vice versa, indicating a potential bidirectional relationship. This review explores the mechanisms and clinical implications of this interplay and proposes integrated screening and care strategies to improve the quality of life for individuals with both conditions. Full article
(This article belongs to the Special Issue Gluten-Free Food and Celiac Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Overlap mechanisms for developing celiac disease and eating disorders.</p>
Full article ">
Back to TopTop