151
|
Ebersole JL, Kirakodu SS, Zhang XD, Dawson D, Miller CS. Salivary features of periodontitis and gingivitis in type 2 diabetes mellitus. Sci Rep 2024; 14:30649. [PMID: 39730430 DOI: 10.1038/s41598-024-77434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/22/2024] [Indexed: 12/29/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with cellular abnormalities, tissue and organ dysfunctions, and periodontitis. This investigation examined the relationship between the oral microbiome and salivary biomarkers in T2DM patients with or without periodontitis. This cohort (35-80 years) included systemically healthy non-periodontitis (NP; n = 31), T2DM without periodontitis (DWoP; n = 32) and T2DM with periodontitis (DWP; n = 29). The oral microbiome [Operational Taxonomic Units (OTUs)] (16 s rRNA sequencing) and targeted host salivary biomarkers (immunoassays) were assessed. We identified 47 OTUs that were significantly different in abundance between NP samples and any disease subset or between disease subgroups. The most unique microbiome patterns were observed in the DWP group. Differences in genera/species abundance were also observed when T2DM patients were stratified by extent of periodontal inflammation and disease (i.e., generalized versus localized gingivitis/periodontitis). Salivary biomarkers showed significant elevations in MMP-8, MMP-9, resistin, IL-1β, IL-6, IFNα, and BAFF (THFSR13b) comparing generalized to localized periodontitis. Salivary analytes showed significant positive correlations with specific microbiome members, predominantly in DWP patients. Odds ratio analyses reinforced that a panel of biologic markers (IL-6, MMP-8) and bacteria (e.g., Bacteroidetes, Fusobacteria, Spirochaetes) discriminated the severity and extent of periodontal disease in this diabetic population.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, 89131, USA.
| | - Sreenatha S Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Xiaohua D Zhang
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Dolph Dawson
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
- Department of Oral Health Practice, Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Craig S Miller
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
- Department of Oral Health Practice, Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
152
|
Dourdouna MM, Kourlaba G, Michos A. QuantiFERON SARS-CoV-2 assay for the evaluation of cellular immunity after immunization with mRNA SARS-CoV-2 vaccines: a systematic review and meta-analysis. Immunol Res 2024; 73:25. [PMID: 39729138 DOI: 10.1007/s12026-024-09570-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/21/2024] [Indexed: 12/28/2024]
Abstract
A systematic review and meta-analysis were performed to evaluate the virus-specific T-cell response after COVID-19 mRNA vaccination, using the QuantiFERON SARS-CoV-2 interferon-γ release assay. A search was conducted (June 8, 2023) in the PUBMED, SCOPUS, and medRxiv databases, to identify studies reporting the QuantiFERON SARS-CoV-2 (Starter (two antigen tubes) or Starter + Extended Pack (three antigen tubes), cut-off ≥ 0.15 IU/mL) positivity rate (PR) in immunocompetent adults, following the administration of two or three COVID-19 mRNA vaccine doses. Study quality was evaluated with the Critical Appraisal Skills Programme Tool. A meta-analysis was conducted using a random-effects model. Heterogeneity and publication bias were assessed. Eleven eligible studies (with 5-73 vaccinated immunocompetent participants) were identified. For COVID-19-naïve participants, ≤ 3 months after the second dose, the pooled PR (random-effects model) was 86 (95% confidence interval (95% CI) 78-95%). Comparing the Starter vs. the Starter + Extended Pack, a significant difference in PRs was detected (80.6% vs. 100% p-value < 0.001). At 3-6 and >6 months after the second dose and ≥ 3 months after the third dose, the pooled PRs were 59% (95% CI 45-72%), 79% (95% CI 66-92%), and 66% (95% CI 50-82%), respectively. For convalescent participants, ≥ 6 months after the third dose, the pooled PR was 81% (95% CI 67-95%). Limitations include heterogeneity and a small number of studies, at some timepoints. In conclusion, following the second or third COVID-19 mRNA vaccine dose, QuantiFERON SARS-CoV-2 detected positive responses in a certain percentage of the vaccinees, possibly because of waning immunity, reduced assay sensitivity, or lack of T-cell response induction in some vaccinees. The detection of positive responses was higher when the Starter + Extended Pack was used. PROSPERO Registration Number: CRD42023431315.
Collapse
Affiliation(s)
- Maria-Myrto Dourdouna
- Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, Aghia Sophia" Children's Hospital, 11527, Athens, Greece
| | - Georgia Kourlaba
- Department of Nursing, University of Peloponnese, Tripoli, Greece
| | - Athanasios Michos
- Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, Aghia Sophia" Children's Hospital, 11527, Athens, Greece.
| |
Collapse
|
153
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
154
|
Rajan A, L K, S V, K S S, M D H, S D, K M, Raveendran R, P V A, T M M, T S, D DR. Correlation of inflammatory markers and NFATC4 gene expression among subjects with prediabetes. J Cardiovasc Thorac Res 2024; 16:264-274. [PMID: 40027364 PMCID: PMC11866773 DOI: 10.34172/jcvtr.33272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/10/2024] [Indexed: 03/05/2025] Open
Abstract
Introduction Prediabetes, characterized by mildly elevated blood sugar levels, significantly increases the risk of developing type 2 diabetes and cardiovascular disease. The condition is linked to higher levels of IL-18, TNF-α, and IL-6, indicating inflammation that may drive type 2 Diabetes Mellitus (T2DM). Despite the known role of inflammation in glucose homeostasis, the involvement of the Nuclear Factor of Activated T Cells 4 (NFATC4) gene in prediabetes remains underexplored. This case-control study aims to investigate the association between physiological, demographic, anthropometric, lifestyle factors, inflammatory markers and NFATC4 gene expression, in the context of prediabetes. Methods The study involved 300 participants aged 20 to 50, with 150 diagnosed with prediabetes and 150 healthy controls. After obtaining informed consent fasting venous blood samples were collected for comprehensive assessments, including biochemical, endocrinological and immunological analyses. Specifically, NFATC4 gene expression and inflammatory markers were measured. Results The findings revealed significantly elevated levels of IL-18, TNF-α, IL-6, and NFATC4 expression in prediabetic individuals compared to controls. Notably, strong positive correlations were observed between NFATC4 expression and the inflammatory markers. Receiver operating characteristic (ROC) curve analysis identified IL-18 and NFATC4 as the most promising biomarkers for predicting prediabetes, followed by TNF-α and IL-6. Multivariate regression analysis further identified socioeconomic status (SES), IL-18, NFATC4, TSH, triglycerides, and HDL as independent predictors of prediabetes. Conclusion These results highlight the key role of inflammation and NFATC4 in prediabetes, stressing the need for strategies to prevent progression to type 2 diabetes and cardiovascular issues.
Collapse
Affiliation(s)
- Aswathi Rajan
- Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam, Chennai, Tamil Nadu, India
| | - Karpagavel L
- Department of Biochemistry, Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam, Chennai, Tamil Nadu, India
| | - Vidya S
- Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam, Chennai, Tamil Nadu, India
| | - Sheena K S
- Chettinad Academy of Research and Education (Deemed to be University), Kelambakkam, Chennai, Tamil Nadu, India
| | - Harilal M D
- Department of Anatomy, Sri Sankara Dental College, Akathumuri P. O, Kerala, India
| | - Deepthi S
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Manjusha K
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Rachana Raveendran
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Ambili P V
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Midhun T M
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Swathi T
- Meenakshi Academy of Higher Education and Research (MAHER- Deemed to be University), West K.K Nagar, Chennai, Tamil Nadu, India
| | - Dinesh Roy D
- Genetika, Centre for Advanced Genetic Studies, Thiruvananthapuram – 695024, Kerala, India
| |
Collapse
|
155
|
Sun G, Ma X, Xu S, Su B, Chen Q, Dong X, Wang L, Wan J, Shi H. Mediation role of body mass index in the relationship between food-specific serum immunoglobulin G reactivity and colorectal adenomas in a Chinese population: a cross-sectional study. Therap Adv Gastroenterol 2024; 17:17562848241307601. [PMID: 39717539 PMCID: PMC11664519 DOI: 10.1177/17562848241307601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024] Open
Abstract
Background Colorectal adenomas (CAs) represent a significant global health issue, particularly in China, where lifestyle modifications have contributed to their increased prevalence. These adenomas are precursors to colorectal cancer. While high-fiber diets have been shown to decrease risk, the implications of food-specific serum immunoglobulin G reactivity (FSsIgGR) on CAs remain uncertain and warrant further investigation. Objectives To investigate the association between FSsIgGR and the occurrence of CAs in the Chinese population, assess the mediating influence of body mass index (BMI), and offer insights into potential prevention strategies. Design A retrospective cross-sectional study. Methods This study is based on 8796 individuals who underwent colonoscopy at the Second Medical Center of Chinese PLA General Hospital from 2017 to 2021. We examined the relationship between FSsIgGR and CAs using logistic regression, controlling for various confounders. Interaction effects were explored through subgroup analysis. We addressed missing data using multiple imputation and confirmed the robustness of our findings through sensitivity analysis. The role of BMI as a mediator was quantified using structural equation modeling. Results The cohort comprised 2703 patients diagnosed with CAs and 6093 polyp-free controls, with an average age of 50.1 years, of whom 70.1% were male. The analysis revealed a significant inverse association between FSsIgGR and the incidence of CAs (adjusted odds ratio = 0.97; 95% confidence interval: 0.95-0.99; p < 0.001). Dose-response analysis indicated a linear reduction in CAs risk correlating with an increased number of IgG-positive food items. Structural equation modeling showed that BMI mediated 6.02% of the effect on CAs risk (p = 0.038). Conclusion Our findings suggest that FSsIgGR correlates with a reduced risk of developing CAs, with BMI partially mediating this effect. These results add a novel dimension to CAs risk assessment and prevention, highlighting potential dietary interventions.
Collapse
Affiliation(s)
- Guanchao Sun
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaona Ma
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shiping Xu
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatrics Diseases, Beijing, China
| | - Binbin Su
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Geriatrics Diseases, Beijing, China
| | - Qianqian Chen
- Department of Gastroenterology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoyu Dong
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lihui Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jun Wan
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China National Clinical Research Center for Geriatrics Diseases, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Hui Shi
- Department of Gastroenterology, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
- National Clinical Research Center for Geriatrics Diseases, 28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
156
|
Osser G, Osser B, Toth C, Miuța CC, Marconi GR, Bondar LI. Exploring the Relationship Between Ejection Fraction, Arterial Stiffness, NT-proBNP, and Hospitalization Risk in Heart Failure Patients. Diagnostics (Basel) 2024; 14:2885. [PMID: 39767246 PMCID: PMC11675150 DOI: 10.3390/diagnostics14242885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/11/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Heart failure (HF) remains a leading cause of hospitalization and morbidity. Arterial stiffness, measured by pulse wave velocity (PWV) and the augmentation index (AIx), has been linked to HF severity and prognosis. This study investigates the relationship between clinical parameters, biochemical indicators, and arterial stiffness in hospitalized patients with HF, aiming to identify predictors of hospitalization and improve patient management. Methods: This cross-sectional study included 98 patients admitted with HF: 53 with acutely decompensated HF (sudden worsening of symptoms) and 45 with chronic HF (stable symptoms of HF). Clinical and biochemical parameters, including ejection fraction (EF), N-terminal prohormone of brain natriuretic peptide (NT-proBNP) levels, and arterial stiffness indicators (PWV and AIx), were measured at admission. During follow-up, 59 patients required re-hospitalization due to acutely decompensated HF, while 39 remained outpatients without further hospitalization. The relationship between these parameters was analyzed using Pearson correlation coefficients, and multiple Cox regression analysis was conducted to identify independent predictors of re-hospitalization. Results: A significant negative correlation between EF and PWV was found (r = -0.853, 95% CI [-0.910, -0.764]), suggesting an association between improved heart function (higher EF) and reduced arterial stiffness (lower PWV). A moderate positive correlation between EF and AIx (r = 0.626, 95% CI [0.473, 0.805]) suggests that, while higher EF is associated with increased AIx, the relationship is weaker compared to EF and PWV. This may reflect differing contributions of vascular and myocardial factors to HF severity. Hospitalized patients exhibited significantly poorer clinical and biochemical profiles, including higher NT-proBNP levels (p < 0.001) and worse blood pressure (BP) measurements (systolic and diastolic, p < 0.01). Multiple Cox regression analysis identified PWV, Aix, and NT-proBNP as independent predictors of re-hospitalization in HF patients, with significant hazard ratios: PWV (HR = 1.15, p = 0.02), AIx (HR = 1.03, p = 0.02), and NT-proBNP (HR = 1.0001, p < 0.01). Conclusions: Arterial stiffness indices (PWV and AIx), EF, and NT-proBNP were identified as significant predictors of re-hospitalization in HF patients. These findings suggest that integrating arterial stiffness measurements into routine clinical assessments may enhance the risk stratification and inform targeted interventions to reduce hospitalizations and improve outcomes.
Collapse
Affiliation(s)
- Gyongyi Osser
- Faculty of Physical Education and Sport, “Aurel Vlaicu” University of Arad, 310130 Arad, Romania; (G.O.); (B.O.); (C.T.); (G.R.M.)
| | - Brigitte Osser
- Faculty of Physical Education and Sport, “Aurel Vlaicu” University of Arad, 310130 Arad, Romania; (G.O.); (B.O.); (C.T.); (G.R.M.)
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Csongor Toth
- Faculty of Physical Education and Sport, “Aurel Vlaicu” University of Arad, 310130 Arad, Romania; (G.O.); (B.O.); (C.T.); (G.R.M.)
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania;
| | - Caius Calin Miuța
- Faculty of Physical Education and Sport, “Aurel Vlaicu” University of Arad, 310130 Arad, Romania; (G.O.); (B.O.); (C.T.); (G.R.M.)
| | - Gabriel Roberto Marconi
- Faculty of Physical Education and Sport, “Aurel Vlaicu” University of Arad, 310130 Arad, Romania; (G.O.); (B.O.); (C.T.); (G.R.M.)
| | - Laura Ioana Bondar
- Doctoral School of Biomedical Sciences, University of Oradea, 410087 Oradea, Romania;
- Department of Biology and Life Sciences, “Vasile Goldiș” Western University of Arad, 310048 Arad, Romania
| |
Collapse
|
157
|
Zeng G, Lu X, Li P, Zeng T, Lin Z, Miao Y, Yuan S, Liu X, Zeng L. Is it feasible to treat polycystic ovarian syndrome with or without insulin resistance using glucokinase activators as novel hypoglycaemic medications? A protocol for a systematic review and meta-analysis. BMJ Open 2024; 14:e088484. [PMID: 39806679 PMCID: PMC11667369 DOI: 10.1136/bmjopen-2024-088484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
INTRODUCTION A variety of hypoglycaemic drugs are used to treat polycystic ovarian syndrome (PCOS), but their efficacy remains insufficient. Glucokinase activators (GKAs) are a unique class of hypoglycaemic medications with emerging potential, notably in significantly reducing insulin resistance (IR). Nevertheless, the efficacy of GKAs in treating PCOS, particularly in the absence or presence of IR, remains uncertain. The meta-analysis protocol aims to address this knowledge gap, furnish evidence-based data to support potential revisions in PCOS treatment guidelines and promote the utilisation of GKAs in clinical settings. METHODS AND ANALYSIS A comprehensive search will be conducted across the Cochrane Central Register of Controlled Trials, PubMed, Web of Science, Embase, Medline, Scopus, CNKI, Wanfang and VIP databases to identify randomised controlled trials investigating the use of GKAs in the treatment of PCOS, irrespective of the presence of IR. The search will encompass all available studies without language restrictions and cover the period from the inception of each database to 10 April 2024. Disputes will be resolved by talking with a third expert following the screening of articles and data extraction by two reviewers. The primary outcomes of interest encompass changes in anthropometric parameters, menstrual frequency, sex hormone levels, and glucose metabolism, while secondary objectives include lipid metabolism and adverse events. The methodological quality of each study will be assessed using Version 2 of the Cochrane Collaboration tool for assessing Risk of Bias (RoB 2.0), and the Grade of Recommendations, Assessment, Development and Evaluation (GRADE) technique will be used to assess the quality of evidence and degree of recommendation. The study duration of this study will be from 5 April 2024 to 10 April 2025. ETHICS AND DISSEMINATION Since this study just analyses data that are readily available to the public and does not directly involve patient participation, ethical approval is not necessary. The findings will be made public by being published in a medical journal that is subject to peer review. PROSPERO REGISTRATION NUMBER CRD42024535633.
Collapse
Affiliation(s)
- Genping Zeng
- Guangzhou University of Chinese Medicine First Clinical School of Medicine, Guangzhou, Guangdong, China
| | - Xijing Lu
- Guangzhou University of Chinese Medicine First Clinical School of Medicine, Guangzhou, Guangdong, China
| | - Peiyin Li
- Guangzhou University of Chinese Medicine First Clinical School of Medicine, Guangzhou, Guangdong, China
| | - Tan Zeng
- Yichun University Medical College, Yichun, Jiangxi, China
| | - Zitong Lin
- Guangzhou University of Chinese Medicine First Clinical School of Medicine, Guangzhou, Guangdong, China
| | - Yuxi Miao
- Guangzhou University of Chinese Medicine First Clinical School of Medicine, Guangzhou, Guangdong, China
| | - Shuo Yuan
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiaojing Liu
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Lei Zeng
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
158
|
Melano I, Azamor T, Caetano CC, Meyer NM, Onwubueke C, Visperas A, Familiar-Macedo D, Salem GM, Soos BL, Calabrese CM, Choi YJ, Chen S, Choi Y, Wu X, Vasconcelos Z, Comhair SA, Nielsen-Saines K, Calabrese LH, Husni ME, Jung JU, Piuzzi NS, Foo SS, Chen W. SARS-CoV-2 ORF8 drives osteoclastogenesis in preexisting immune-mediated inflammatory diseases. JCI Insight 2024; 9:e178820. [PMID: 39704167 DOI: 10.1172/jci.insight.178820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 11/05/2024] [Indexed: 12/21/2024] Open
Abstract
Patients with immune-mediated inflammatory diseases (IMIDs) like rheumatoid arthritis (RA) are at higher risk for severe COVID-19 and long-term complications in bone health. Emerging clinical evidence demonstrated that SARS-CoV-2 infection reduces bone turnover and promotes bone loss, but the mechanism underlying worsened bone health remains elusive. This study sought to identify specific immune mediators that exacerbated preexisting IMIDs after SARS-CoV-2 exposure. Plasma samples from 4 groups were analyzed: healthy, IMID only, COVID-19 only, and COVID-19 + IMID. Using high-throughput multiplexed proteomics, we profiled 1,500 protein biomarkers and identified 148 unique biomarkers in COVID-19 patients with IMIDs, including elevated inflammatory cytokines (e.g., IL-17F) and bone resorption markers. Long-term circulating SARS-CoV-2 ORF8, a virulence factor for COVID-19, was detected in the COVID + IMID group. RA was one of the most common IMIDs in our study. ORF8 treatment of RA-derived human osteoblasts (RA-hOBs) increased levels of inflammatory (TNF, IL6, CCL2) and bone resorption (RANKL/osteoprotegerin ratio) markers compared with healthy controls. Supernatants from ORF8-treated RA-hOBs drove the differentiation of macrophages into osteoclast-like cells. These findings suggest that SARS-CoV-2 exposure can exacerbate IMIDs through ORF8-driven inflammation and osteoclastogenesis, highlighting potential therapeutic targets for managing COVID-19-induced bone pathologies.
Collapse
Affiliation(s)
- Ivonne Melano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tamiris Azamor
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Camila Cs Caetano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nikki M Meyer
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Chineme Onwubueke
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
| | | | - Débora Familiar-Macedo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Gielenny M Salem
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Brandy-Lee Soos
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cassandra M Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Youn Jung Choi
- Department of Medicine, Kao Autoimmunity Institute, and Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuyang Chen
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Younho Choi
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, Florida, USA
| | - Xianfang Wu
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
| | | | - Suzy Aa Comhair
- Respiratory Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Karin Nielsen-Saines
- Department of Pediatrics, Division of Pediatric Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Leonard H Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - M Elaine Husni
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jae U Jung
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
| | | | - Suan-Sin Foo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
| | - Weiqiang Chen
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, CWRU School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
159
|
Tiongco RE, Flores JA, Castro EJ, Dayrit SA, Dominguez MJ, Manahan E, Pineda-Cortel MR. Association of Strongyloides stercoralis infection with the development of diabetes mellitus: a meta-analysis. J Helminthol 2024; 98:e86. [PMID: 39703057 DOI: 10.1017/s0022149x24000828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Previous studies have shown that helminth infection protects against the development of diabetes mellitus (DM), possibly related to the hygiene hypothesis. However, studies involving Stronglyoides stercoralis and its possible association with DM are scarce and have shown contradicting results, prompting us to perform this meta-analysis to obtain more precise estimates. Related studies were searched from PubMed, Google Scholar, Science Direct, and Cochrane Library until 1 August 2024. Data on the occurrence of DM in patients positive and negative for S. stercoralis were obtained. All analyses were done using Review Manager 5.4. The initial search yielded a total of 1725 studies, and after thorough screening and exclusion, only five articles involving 2106 participants (536 cases and 1570 controls) were included in the meta-analysis. Heterogeneity was assessed, and outlier studies were excluded using a funnel plot. Results showed a significant association of S. stercoralis infection with DM, suggesting that those with the infection are less likely to develop DM. Overall, the results suggest that S. stercoralis infection may decrease the likelihood of developing DM, potentially supporting the hygiene hypothesis.
Collapse
Affiliation(s)
- R E Tiongco
- Department of Medical Technology, School of Health Sciences, UST General Santos, General Santos City 9500, Philippines
| | - J A Flores
- Department of Medical Technology, College of Allied Medical Professions, Angeles University Foundation, Angeles City2009, Philippines
| | - E J Castro
- Department of Medical Technology, College of Allied Medical Professions, Angeles University Foundation, Angeles City2009, Philippines
| | - S A Dayrit
- Department of Medical Technology, College of Allied Medical Professions, Angeles University Foundation, Angeles City2009, Philippines
| | - M J Dominguez
- School of Medicine, Angeles University Foundation, Angeles City2009, Philippines
| | - E Manahan
- Department of Medical Technology, School of Health Sciences, UST General Santos, General Santos City 9500, Philippines
- Department of Medical Technology, Faculty of Pharmacy, University of Santo Tomas, Manila1008, Philippines
| | - M R Pineda-Cortel
- Department of Medical Technology, Faculty of Pharmacy, University of Santo Tomas, Manila1008, Philippines
- Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila1008, Philippines
| |
Collapse
|
160
|
Xu F, Li X, Wang X, Wu H, Chen S, Chen J, Kong X, Yang Z. Revealing therapeutic targets and drugs from Chinese medicine for ulcerative colitis using bioinformatics. J Biomol Struct Dyn 2024:1-11. [PMID: 39693490 DOI: 10.1080/07391102.2024.2440651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/03/2024] [Indexed: 12/20/2024]
Abstract
Pathogenesis and therapeutic drugs for ulcerative colitis (UC) have plagued researchers worldwide. In this study, therapeutic targets, and drugs from Chinese medicines for UC were screened using bioinformatics. We downloaded five datasets from the GEO database and three machine learning algorithms were used for screening diagnostic biomarkers of UC. Combined with the differential genes for UC, gene sets related to bile acid metabolism, short-chain fatty acids, apoptosis, pyroptosis, G-protein-coupled receptors, mitochondria, and autophagy were collected to screen the core targets, and analyze the association of therapeutic genes (diagnostic biomarkers and core targets) with immune cells. In addition, screening ingredients of Chinese medicines based on UC therapeutic targets was performed. Molecular docking, molecular dynamics simulation, and literature validation were also performed. The screening yielded 37 key therapeutic targets, including 5 diagnostic biomarkers (CCL11, CXCL1, PDZK1IP1, TIMP1, and UGT2A3) and 32 core targets based on hot gene sets. Immune cell infiltration was strongly associated with therapeutic targets in UC, especially neutrophils, macrophages, mast cells, and dendritic cells. Furthermore, a total of 33 compounds with high safety had been recognized as having potential to mitigate UC by reverse prediction from Chinese medicines, and molecular docking, molecular dynamics simulation, and literature reports preliminarily validated the screening results. Although further experimental validation is needed, this work provides some potential therapeutic targets and drugs from Chinese medicines against UC.
Collapse
Affiliation(s)
- Feng Xu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaofen Li
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiangpei Wang
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang, China
| | - Hongmei Wu
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Song Chen
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jianyang Chen
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiangxi Kong
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhenglin Yang
- Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
161
|
Yang F, Zhao LJ, Xu Q, Zhao J. The journey of p38 MAP kinase inhibitors: From bench to bedside in treating inflammatory diseases. Eur J Med Chem 2024; 280:116950. [PMID: 39406118 DOI: 10.1016/j.ejmech.2024.116950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/06/2024] [Indexed: 11/25/2024]
Abstract
The p38 mitogen-activated protein kinase (MAPK) pathway is pivotal in regulating inflammatory responses and has emerged as a key target for the development of small-molecule inhibitors aimed at treating inflammatory diseases. In arthritis, especially rheumatoid arthritis (RA), the p38 MAPK pathway contributes to chronic inflammation and joint destruction by promoting the production of pro-inflammatory cytokines. Preclinical studies have shown that small-molecule inhibitors targeting the p38 MAPK pathway hold significant promise, exhibiting the potential to reduce inflammation and preserve joint integrity. Targeting this pathway presents a novel therapeutic approach to mitigating inflammation. This review traces the evolution of p38 MAP kinase inhibitors from initial laboratory studies to clinical candidates, underscoring their potential to significantly alter the treatment approach for inflammatory diseases.
Collapse
Affiliation(s)
- Fuwei Yang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Li-Jie Zhao
- The Rogel Cancer Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, United States.
| | - Qinli Xu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Jianhui Zhao
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
162
|
Hernyák M, Tóth LI, Csiha S, Molnár Á, Lőrincz H, Paragh G, Harangi M, Sztanek F. Kallistatin as a Potential Marker of Therapeutic Response During Alpha-Lipoic Acid Treatment in Diabetic Patients with Sensorimotor Polyneuropathy. Int J Mol Sci 2024; 25:13276. [PMID: 39769041 PMCID: PMC11675709 DOI: 10.3390/ijms252413276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic sensorimotor neuropathy (DSPN) is strongly associated with the extent of cellular oxidative stress and endothelial dysfunction in type 2 diabetes (T2DM). Alpha-lipoic acid (ALA) attenuates the progression of DSPN through its antioxidant and vasculoprotective effects. Kallistatin has antioxidant and anti-inflammatory properties. We aimed to evaluate changes in kallistatin levels and markers of endothelial dysfunction in patients with T2DM and DSPN following six months of treatment with 600 mg/day of ALA. A total of 54 patients with T2DM and DSPN and 24 control patients with T2DM but without neuropathy participated in this study. The serum concentrations of kallistatin, ICAM-1, VCAM-1, oxLDL, VEGF, ADMA, and TNF-alpha were measured by an ELISA. Peripheral sensory neuropathy was assessed with neuropathy symptom questionnaires and determination of the current perception threshold. After ALA treatment, the level of kallistatin significantly decreased, as well as the levels of TNF-alpha and ADMA. Changes in kallistatin levels were positively correlated with changes in oxLDL. The improvement in DSPN symptoms following ALA treatment showed a positive correlation with changes in kallistatin, VEGF, oxLDL, and ADMA levels. Based on our results, kallistatin could represent a potential new biomarker for assessing therapeutic response during ALA treatment in patients with DSPN.
Collapse
Affiliation(s)
- Marcell Hernyák
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Doctoral School of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
| | - László Imre Tóth
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Doctoral School of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
| | - Sára Csiha
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Ágnes Molnár
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Hajnalka Lőrincz
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - György Paragh
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| | - Mariann Harangi
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
- Institute of Health Studies, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary
- ELKH-UD Vascular Pathophysiology Research Group 11003, University of Debrecen, H-4032 Debrecen, Hungary
| | - Ferenc Sztanek
- Division of Metabolism, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (M.H.); (L.I.T.); (S.C.); (Á.M.); (H.L.); (G.P.); (M.H.)
| |
Collapse
|
163
|
Apte A, Dutta Dey P, Julakanti SR, Midura-Kiela M, Skopp SM, Canchis J, Fauser T, Bardill J, Seal S, Jackson DM, Ghishan FK, Kiela PR, Zgheib C, Liechty KW. Oral Delivery of miR146a Conjugated to Cerium Oxide Nanoparticles Improves an Established T Cell-Mediated Experimental Colitis in Mice. Pharmaceutics 2024; 16:1573. [PMID: 39771552 PMCID: PMC11679827 DOI: 10.3390/pharmaceutics16121573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Dysregulated inflammation and oxidative stress are strongly implicated in the pathogenesis of inflammatory bowel disease. We have developed a novel therapeutic that targets inflammation and oxidative stress. It is comprised of microRNA-146a (miR146a)-loaded cerium oxide nanoparticles (CNPs) (CNP-miR146a). We hypothesized that oral delivery of CNP-miR146a would reduce colonic inflammation in a mouse model of established, chronic, T cell-mediated colitis. Methods: The stability of CNP-miR146a and mucosal delivery was assessed in vitro with simulated gastrointestinal fluid and in vivo after oral gavage by quantitative real-time RT-PCR. The efficacy of orally administered CNP-miR146a was tested in mice with established colitis using the model of adoptive naïve T-cell transfer in recombinant activating gene 2 knockout (Rag2-/-) mice. Measured outcomes included histopathology; CD45+ immune cell infiltration; oxidative DNA damage (tissue 8-hydroxy-2'-deoxyguanosine; 8-OHdG); expression of IL-6 and TNF mRNA and protein, and flow cytometry analysis of lamina propria Th1 and Th17 cell populations. Results: miR146a expression remained stable in simulated gastric and intestinal conditions. miR146a expression increased in the intestines of mice six hours following oral gavage of CNP-miR146a. Oral delivery of CNP-miR146a in mice with colitis was associated with reduced inflammation and oxidative stress in the proximal and distal colons as evidenced by histopathology scoring, reduced immune cell infiltration, reduced IL-6 and TNF expression, and decreased populations of CD4+Tbet+IFNg+ Th1, CD4+RorgT+IL17+ Th17, as well as pathogenic double positive IFNg+IL17+ T cells. Conclusions: CNP-miR146a represents a novel orally available therapeutic with high potential to advance into clinical trials.
Collapse
Affiliation(s)
- Anisha Apte
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Pujarini Dutta Dey
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Srisaianirudh Reddy Julakanti
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Monica Midura-Kiela
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Stacy M. Skopp
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Jimena Canchis
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - Tobias Fauser
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
| | - James Bardill
- Laboratory for Fetal and Regenerative Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | | | - Fayez K. Ghishan
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Pawel R. Kiela
- Department of Pediatrics, Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children’s Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
- Department of Immunobiology, University of Arizona Health Sciences Center, Tucson, AZ 85621, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| | - Kenneth W. Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Arizona Tucson College of Medicine, Banner Children’s at Diamond Children’s Medical Center, 1656 E Mabel St, Rm 230, Tucson, AZ 85721, USA
- Ceria Therapeutics, Inc., Tucson, AZ 85721, USA
| |
Collapse
|
164
|
Abdollahi A, Nateghi S, Panahi Z, Inanloo SH, Salarvand S, Pourfaraji SM. The association between mortality due to COVID-19 and coagulative parameters: a systematic review and meta-analysis study. BMC Infect Dis 2024; 24:1373. [PMID: 39623325 PMCID: PMC11610108 DOI: 10.1186/s12879-024-10229-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/14/2024] [Indexed: 12/06/2024] Open
Abstract
AIMS AND OBJECTIVES This systematic review and meta-analysis study evaluated the association between mortality due to COVID-19 and coagulative factors. METHODS A systematic search was conducted on electronic databases including PubMed, Scopus, and the Web of Science from the beginning of the pandemic until October 2024 to identify relevant studies on COVID-19 patients and their laboratory findings related to coagulation markers and mortality outcome. Eligibility criteria were defined based on the PICO framework, and data extraction was performed by two authors independently using a standardized sheet. Statistical analysis was accomplished using the random effects model, and heterogeneity among studies was assessed using the I2 test. R and RStudio were used for statistical analysis and visualization. RESULTS Our systematic literature search yielded 6969 studies, with 48 studies meeting the inclusion criteria for our meta-analysis. The mean platelet count was significantly lower in deceased COVID-19 patients compared to survivors (20.58), while activated partial thromboplastin time (aPTT) and fibrinogen levels did not show significant differences. The pooled mean difference of D-Dimer, International Normalized Ratio (INR), and prothrombin time (PT) were significantly lower in survived patients (-2.45, -0.10, and -0.84, respectively). These findings suggest that platelet count, D-Dimer, INR, and PT may serve as potential indicators of mortality in COVID-19 patients. CONCLUSION The results of our systematic review and meta-analysis revealed a significant reduction in the pooled platelet count among deceased individuals when compared to survivors. However, no significant distinctions were observed in the pooled mean activated aPTT and fibrinogen levels between the deceased and survivor groups. On the other hand, there were noticeable variations in the pooled estimated mean of INR, PT, and D-Dimer levels, with significantly higher values in the deceased group compared to those who survived.
Collapse
Affiliation(s)
- Alireza Abdollahi
- Department of Pathology, School of Medicine, IKHC, Teheran University of Medical Sciences, Tehran, Iran
| | - Saeed Nateghi
- Department of Cardiology, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Panahi
- School of Medicine, Department of Obstetrics and Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Inanloo
- Department of Urology, Sina Hospital Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Salarvand
- Department of Pathology, School of Medicine, IKHC, Teheran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
165
|
Yamaguchi M, Yoshiyama T, Maruyama R, Ohashi M, Nishizono S, Kobayashi T, Miyatake M, Sameshima H. Citrus tamurana Hort. ex Tanaka (Hyuganatsu orange)-derived arabinogalactan suppresses bone turnover in postmenopausal women: A randomized placebo-controlled study. J Obstet Gynaecol Res 2024; 50:2299-2308. [PMID: 39419484 DOI: 10.1111/jog.16116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024]
Abstract
AIM To evaluate Hyuganatsu oranges (Citrus tamurana Hort. Ex Tanaka) derived arabinogalactan for bone turnover, we performed a randomized placebo-controlled trial. METHODS Sixty-three postmenopausal women were age-stratified and randomly assigned to receive arabinogalactan-rich hyuganatsu juice (study group) or a placebo drink (control group) for 90 days. We measured blood tartrate-resistant acid phosphatase 5b (TRACP5b), type I procollagen N-terminal propeptide (P1NP), and other bone turnover biomarker levels at baseline, days 45 and 90 (T90) of the intervention, and day 30 of recovery. Cumulative effects were compared between groups using repeated-measures linear mixed model analysis. The primary endpoint was the difference between the pre- and post-intervention TRACP5b and P1NP levels. RESULTS Using repeated measures linear mixed model analysis, the study group had significantly lower TRACP5b and P1NP levels at day 90 than the control group (mean [95% confidence interval]; TRACP5b: 310.0 [269.2-350.9] vs. 386.4 [341.2-431.6] mU/dL; P1NP: 53.7 [48.6-58.7] vs. 70.3 [64.1-76.4] ng/mL), whereas other biomarker levels showed no change. CONCLUSION Arabinogalactan-rich Hyuganatsu juice suppressed bone mineral turnover and potentially improved ovarian hormone deficiency-induced osteoporosis in postmenopausal women.
Collapse
Affiliation(s)
- Masatoshi Yamaguchi
- Department of Obstetrics and Gynecology, University of Miyazaki, Faculty of Medicine, Miyazaki, Japan
| | | | - Ruriko Maruyama
- Department of Obstetrics and Gynecology, University of Miyazaki, Faculty of Medicine, Miyazaki, Japan
| | - Masanao Ohashi
- Department of Obstetrics and Gynecology, Kawakita General Hospital, Tokyo, Japan
| | - Shoko Nishizono
- Faculty of Biotechnology and Life Science, Sojo University, Kumamoto, Japan
| | - Taichi Kobayashi
- Center for Collaborative Research & Community Cooperation, University of Miyazaki, Miyazaki, Japan
| | - Munetoshi Miyatake
- Faculty of Engineering, University of Miyazaki, University of Miyazaki, Miyazaki, Japan
| | - Hiroshi Sameshima
- Department of Obstetrics and Gynecology, University of Miyazaki, Faculty of Medicine, Miyazaki, Japan
| |
Collapse
|
166
|
Zhang Y, Li B, Fu Y, Cai H, Zheng Y. Txnip promotes autophagic apoptosis in diabetic cardiomyopathy by upregulating FoxO1 and its acetylation. Cell Signal 2024; 124:111469. [PMID: 39396562 DOI: 10.1016/j.cellsig.2024.111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Autophagy dysfunction and apoptosis exacerbate the risk of heart failure in patients with diabetic cardiomyopathy (DCM). However, the interactions between autophagy and apoptosis in DCM and their underlying mechanisms remain poorly understood. This study induced type 1 DCM in C57BL/6 mice via streptozotocin injection and exposed H9C2 cells to high glucose to investigate these mechanisms. The study revealed a significant elevation in autophagic vesicles and compromised autophagic flux, accompanied by pronounced myocardial cell apoptosis in the myocardium of diabetic mice. Long-term exposure to high glucose in H9C2 cells led to enhanced autophagosome formation and impaired autophagic flux, while inhibition of autophagy with 3-MA reduced cell apoptosis. Additionally, we observed an increase in Txnip expression in the myocardium of diabetic mice and in high glucose-treated H9C2 cells, which regulates autophagic apoptosis in high glucose-treated H9C2 cells. Furthermore, Txnip regulates autophagic apoptosis through the modulation of forkhead box-1 (FoxO1) expression and acetylation. Prolonged high glucose exposure resulted in increased levels of phosphorylated sirtuin 1 (SIRT1) and reduced SIRT1/FoxO1 interaction, changes that were ameliorated by Txnip knockdown. Txnip overexpression elevated FoxO1 levels, which could be suppressed by NAC and GSH. These findings revealed that Txnip mediates autophagic apoptosis in DCM by upregulating FoxO1 via ROS and enhancing FoxO1 acetylation through the suppression of SIRT1 activity. The discovery of this new mechanism provides new perspectives and potential therapeutic targets for understanding and treating DCM.
Collapse
Affiliation(s)
- Yaoting Zhang
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Bing Li
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yu Fu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - He Cai
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yang Zheng
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
167
|
Arcieri M, Abrami C, Graziano A, Restaino S, Barbui E, Rizzante E, D'Ippolito S, Vizzielli G, Driul L. The influence of celiac disease on fertility and pregnancy: an Italian survey. Arch Gynecol Obstet 2024; 310:2907-2914. [PMID: 39499311 DOI: 10.1007/s00404-024-07781-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/08/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND Celiac disease (CD) is an inflammatory enteropathy that has been associated to obstetric and gynecological disorders. However, it is still not adequately tested by gynecologists due to the misconception that it is solely a gastrointestinal disease. This underestimation requires the development of targeted interventions. PURPOSE This study aims to evaluate the association between CD and obstetric/gynecological complications, highlight the importance of informing patients about CD manifestations, and assess the patient satisfaction with the information provided by healthcare professionals on the disease. METHODS A digital survey was administered to celiac women via the Italian Celiac Association's website. RESULTS We analyzed 493 questionnaires. Obstetric and gynecological disorders led to the diagnosis of CD in 11.7% of interviewed. The study revealed that untreated CD patients are more predisposed to miscarriages (41.8% vs 34% before/after diagnosis, respectively, p = 0.111), anemia in pregnancy (71.4% vs 40.4% before/after diagnosis, respectively, p < 0.001) and the risk of low birth weight (newborns weighing < 1500 g were 4.0% before and 1.1% after the gluten-free diet, p = 0.028). Women with CD, both before and after gluten-free diet, had higher infertility rates (about 19%) than the general population. Additionally, 73% of interviewees were dissatisfied with the information they received from health professionals about the reproductive implications of CD. CONCLUSION Our research contributes to a deeper understanding of the intersection between CD and reproductive outcomes, highlighting the main obstetric and gynecological problems related to it. It emphasizes the importance of patient's perspective and the need for greater awareness about celiac disease from healthcare workers.
Collapse
Affiliation(s)
- Martina Arcieri
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Carlotta Abrami
- Medical Area Department (DAME), University of Udine, Udine, Italy
| | | | - Stefano Restaino
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy.
- PhD School in Biomedical Sciences, Gender Medicine, Child and Women Health, University of Sassari, Sassari, Italy.
| | - Elisa Barbui
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Elisa Rizzante
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Silvia D'Ippolito
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.), Rome, Italy
| | - Giuseppe Vizzielli
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
- Medical Area Department (DAME), University of Udine, Udine, Italy
| | - Lorenza Driul
- Clinic of Obstetrics and Gynecology, "S. Maria della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
- Medical Area Department (DAME), University of Udine, Udine, Italy
| |
Collapse
|
168
|
Day J, Kushlaf H, Tarvin S, Cavagna L, Codullo V, Shinjo SK, Lyu X, Knitza J, Hajji R, Blier PR, Tseng CW, Appenzeller S, Rider LG, Christopher-Stine L, Gupta L. Telemedicine for the Care of Patients with Idiopathic Inflammatory Myopathies: Experience, Insights and Future Directions from the International Myositis Assessment and Clinical Studies Telemedicine Scientific Interest Group. Curr Rheumatol Rep 2024; 26:414-420. [PMID: 39215900 DOI: 10.1007/s11926-024-01163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Jessica Day
- Department of Rheumatology, Royal Melbourne Hospital, Parkville, Australia
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Hani Kushlaf
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stacey Tarvin
- Riley Hospital for Children at Indiana University, Indianapolis, IN, USA
| | - Lorenzo Cavagna
- Dipartimento di Medicina Interna e Terapia Medica, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico S- Matteo, Pavia, Italy
| | - Veronica Codullo
- Dipartimento di Medicina Interna e Terapia Medica, Università di Pavia, Pavia, Italy
- Division of Rheumatology, Fondazione IRCCS Policlinico S- Matteo, Pavia, Italy
| | - Samuel Katsuyuki Shinjo
- Division of Rheumatology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Xia Lyu
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Johannes Knitza
- Institute of Digital Medicine, University Hospital Giessen-Marburg, Philipps University, Marburg, Germany
| | - Raouf Hajji
- Department of Internal Medicine, Sidi Bouzid Hospital, Ibn Aljazzar Medicine, Faculty of Sousse, Sousse, Tunisia
| | - Peter R Blier
- Department of Paediatrics, Tufts University School of Medicine, Boston, MA, USA
| | - Chih Wei Tseng
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Simone Appenzeller
- Department of Orthopedics, Rheumatology and Traumatology, School of Medical Science, University of Campinas, Campinas, Brazil
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | | | - Latika Gupta
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, UK.
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, WV10 0QP, United Kingdom.
| |
Collapse
|
169
|
Yuan L, Zhou Y, Wang R, Huang X, Tang R, Yan F. Unveiling the role of sTLR2: A novel biomarker for predicting septic-associated AKI. Cytokine 2024; 184:156798. [PMID: 39488192 DOI: 10.1016/j.cyto.2024.156798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) is common in septic patients and strongly associated with adverse outcomes. The pathophysiology of AKI in septic patients remains elusive, and detection of patients at risk of AKI or at risk of progression to severe and persistent AKI is critical for timely and adequate support measures, including mitigating further renal damage. Therefore, identification of biomarkers associated with septic-associated AKI that contribute to improve septic AKI is an area of intensive research. METHODS A total of 116 consecutive patients with sepsis were categorized into two groups (AKI and non-AKI) based on the occurrence of AKI within 24 h of admission to the intensive care unit (ICU). Serum levels of soluble TLR2 (sTLR2), as well as biomarkers such as interleukin(IL)-6, IL-22, IL-10, creatinine, urea, procalcitonin, hypersensitive C-reactive protein (hs-CRP), and D-Dimer (D2), were measured within 24 h after ICU admission. Demographic and clinical characteristics including sequential organ failure assessment (SOFA) scores and acute physiology and chronic health evaluation II (APACHE II) scores were recorded. Logistic regression analysis was conducted to identify potential predictive biomarkers. Receiver operating characteristic (ROC) curve analysis was employed to determine the optimal model for predicting septic-associated AKI. RESULTS Patients in the AKI group exhibited significantly higher serum concentrations of IL-6, IL-10, sTLR2, creatinine, urea, hs-CRP, procalcitonin, D2 and lower serum albumin concentrations as well as higher APACHE II scores compared to those in the non-AKI group. Logistic regression analysis revealed that APACHE II scores, log10-transformed sTLR2 concentration, creatinine and D2 concentration were valuable predictors of AKI among septic patients. ROC curves demonstrated that log10-transformed sTLR2 concentration exhibited comparable predictive value to creatinine in determining the incidence of sepsis-associated AKI. The model with variables of APACHE II score, Log10-transformed serum TLR2 concentration, creatinine and D2 concentration yielded the greatest area under the curve of 0.863. CONCLUSION Elevated levels of sTLR2 in early-stage of septic patients may serve as a promising novel biomarker for predicting sepsis-associated AKI.
Collapse
Affiliation(s)
- Li Yuan
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Yongshuang Zhou
- West China School of Medicine, West China Hospital Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyu Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Department of Organ Transplantation, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, China
| | - Xin Huang
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Ruilin Tang
- Geriatric Diseases Institute of Chengdu, Department of Clinical Laboratory, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China
| | - Fang Yan
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Geriatric Diseases Institute of Chengdu, Department of Intensive Care Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China; Center for Medicine Research and Translation, Chengdu Fifth People's Hospital, Chengdu, Sichuan Province, China.
| |
Collapse
|
170
|
van Dam KPJ, Wieske L, Stalman EW, Kummer LYL, van der Kooi AJ, Raaphorst J, van de Beek D, Verschuuren JJGM, Ruiter AM, Brusse E, van Doorn PA, Baars AE, van der Pol WL, Goedee HS, ten Brinke A, van Ham SM, Rispens T, Kuijpers TW, Eftimov F, the T2B! Immunity Against SARS‐CoV‐2 Study Group. Patient-reported daily functioning after SARS-CoV-2 vaccinations in autoimmune neuromuscular diseases. Eur J Neurol 2024; 31:e16409. [PMID: 39236312 PMCID: PMC11555150 DOI: 10.1111/ene.16409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND AND PURPOSE There are concerns for safety regarding SARS-CoV-2 vaccines for patients with autoimmune neuromuscular disease. We compared daily functioning using disease-specific patient-reported outcome measures (PROMs) before and after SARS-CoV-2 vaccinations. METHODS In this substudy of a prospective observational cohort study (Target-to-B!), patients with myasthenia gravis (MG), chronic inflammatory demyelinating polyneuropathy (CIDP), multifocal motor neuropathy (MMN), and idiopathic inflammatory myopathy (IIM) vaccinated against SARS-CoV-2 were included. Surveys of daily functioning (Myasthenia Gravis Activities of Daily Living, Inflammatory Rasch-Built Overall Disability Scale, Multifocal Motor Neuropathy Rasch-Built Overall Disability Scale, and Health Assessment Questionnaire-Disability Index) were sent before first vaccination and every 60 days thereafter for up to 12 months. Regression models were constructed to assess differences in PROM scores related to vaccination, compared to scores unrelated to vaccination. We also assessed the proportion of patients with deterioration of at least the minimal clinically important difference (MCID) between before first vaccination and 60 days thereafter. RESULTS We included 325 patients (median age = 59 years, interquartile range = 47-67, 156 [48%] female sex), of whom 137 (42%) had MG, 79 (24%) had CIDP, 43 (13%) had MMN, and 66 (20%) had IIM. PROM scores related to vaccination did not differ from scores unrelated to vaccination. In paired PROMs, MCID for deterioration was observed in three of 49 (6%) MG patients, of whom none reported a treatment change. In CIDP, MCID for deterioration was observed in eight of 29 patients (28%), of whom two of eight (25%) reported a treatment change. CONCLUSIONS SARS-CoV-2 vaccination had no effect on daily functioning in patients with autoimmune neuromuscular diseases, confirming its safety in these patients.
Collapse
Affiliation(s)
- Koos P. J. van Dam
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Department of Clinical NeurophysiologySt. Antonius HospitalNieuwegeinthe Netherlands
| | - Eileen W. Stalman
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Laura Y. L. Kummer
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
- Sanquin Research and Landsteiner Laboratory, Department of ImmunopathologyAmsterdam UMCAmsterdamthe Netherlands
| | - Anneke J. van der Kooi
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Joost Raaphorst
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Diederik van de Beek
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | | | - Annabel M. Ruiter
- Department of NeurologyLeiden University Medical CenterLeidenthe Netherlands
| | - Esther Brusse
- Department of NeurologyErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Pieter A. van Doorn
- Department of NeurologyErasmus MC University Medical CenterRotterdamthe Netherlands
| | - Adája E. Baars
- Department of NeurologyErasmus MC University Medical CenterRotterdamthe Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and NeurosurgeryBrain Center UMC UtrechtUtrechtthe Netherlands
| | - H. Stephan Goedee
- Department of Neurology and NeurosurgeryBrain Center UMC UtrechtUtrechtthe Netherlands
| | - Anja ten Brinke
- Department of Neurology and NeurosurgeryBrain Center UMC UtrechtUtrechtthe Netherlands
| | - S. Marieke van Ham
- Sanquin Research and Landsteiner Laboratory, Department of ImmunopathologyAmsterdam UMCAmsterdamthe Netherlands
- Swammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamthe Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Department of ImmunopathologyAmsterdam UMCAmsterdamthe Netherlands
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology, and Infectious Disease, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Filip Eftimov
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamthe Netherlands
| | | |
Collapse
|
171
|
Xu J, Li M, Jiang X, Wang Y, Ma H, Zhou Y, Tian M, Liu Y. Omentin-1 and diabetes: more evidence but far from enough. Arch Physiol Biochem 2024; 130:599-605. [PMID: 37395595 DOI: 10.1080/13813455.2023.2230380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 07/04/2023]
Abstract
AIMS AND BACKGROUND Omentin-1 (oment-1) is a type of adipokines that is mainly expressed in visceral fat tissue. Based on accumulating evidence, oment-1 is closely related to diabetes and its complications. However, so far data about oment-1 and diabetes is fragmented. In this review, we focus on the role of oment-1 on diabetes, including its possible signalling pathways, the correlation of circulating omens-1 levels with diabetes and its complications. METHODS The web of PubMed was searched for articles of relevant studies published until February, 2023. RESULTS AND CONCLUSIONS Oment-1 might exert its effects by inhibiting the NF-κB pathway and activating the Akt and AMPK-dependent pathways. The level of circulating oment-1 is negatively correlated with the occurrence of type 2 diabetes and some complications, including diabetic vascular disease, cardiomyopathy, and retinopathy, which can be affected by anti-diabetic therapies. Oment-1 could be a promising marker for screening and targeted therapy for diabetes and its complications; however, more studies are still needed.
Collapse
Affiliation(s)
- Jing Xu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Min Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinli Jiang
- Department of Ophthalmology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuling Wang
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang City, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, China
| | - Yaru Zhou
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Meimei Tian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
172
|
Qadir MMF, Elgamal RM, Song K, Kudtarkar P, Sakamuri SSVP, Katakam PV, El-Dahr SS, Kolls JK, Gaulton KJ, Mauvais-Jarvis F. Sex-specific regulatory architecture of pancreatic islets from subjects with and without type 2 diabetes. EMBO J 2024; 43:6364-6382. [PMID: 39567827 PMCID: PMC11649919 DOI: 10.1038/s44318-024-00313-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
Patients with type 2 and type 1 diabetes (T2D and T1D) exhibit sex-specific differences in insulin secretion, the mechanisms of which are unknown. We examined sex differences in human pancreatic islets from 52 donors with and without T2D combining single cell RNA-sequencing (scRNA-seq) and single nucleus ATAC-sequencing (snATAC-seq) with assays probing hormone secretion and bioenergetics. In non-diabetic (ND) donors, sex differences in islet cell chromatin accessibility and gene expression predominantly involved sex chromosomes. In contrast, islets from T2D donors exhibited similar sex differences in sex chromosome-encoded differentially expressed genes (DEGs) as ND donors, but also exhibited sex differences in autosomal genes. Comparing β cells from T2D and ND donors, gene enrichment of female β cells showed suppression in mitochondrial respiration, while male β cells exhibited suppressed insulin secretion, suggesting a role for mitochondrial failure in females in the transition to T2D. We finally performed cell type-specific, sex stratified, GWAS restricted to differentially accessible chromatin peaks across T2D, fasting glucose, and fasting insulin traits. We identified that differentially accessible regions overlap with T2D-associated variants in a sex- and cell type-specific manner.
Collapse
Affiliation(s)
- Mirza Muhammad Fahd Qadir
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA
- Tulane Center of Excellence in Sex-Based Precision Medicine, New Orleans, LA, USA
| | - Ruth M Elgamal
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Kejing Song
- Center for Translational Research in Infection and Inflammation, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Parul Kudtarkar
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Prasad V Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Samir S El-Dahr
- Department of Pediatrics, Tulane University, School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kyle J Gaulton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
- Tulane Center of Excellence in Sex-Based Precision Medicine, New Orleans, LA, USA.
| |
Collapse
|
173
|
Dey M, Doskaliuk B, Parodis I, Lindblom J, Wincup C, Joshi M, Dey D, Katchamart W, Kadam E, Sen P, Shinjo SK, Nune A, Goo PA, Ziade N, Chen YM, Traboco LS, Gutiérrez CET, Vaidya B, Agarwal V, Gupta L, Nikiphorou E. COVID-19 vaccination-related delayed adverse events among people with rheumatoid arthritis: results from the international COVAD survey. Rheumatol Int 2024; 44:2853-2861. [PMID: 39503760 PMCID: PMC11618185 DOI: 10.1007/s00296-024-05742-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 10/14/2024] [Indexed: 12/03/2024]
Abstract
This study aimed to assess COVID-19 vaccination-related AEs in patients with rheumatoid arthritis (RA), in the COVID-19 Vaccination in Autoimmune Diseases (COVAD)-2 study. An online international cross-sectional survey captured self-reported data on COVID-19 vaccination-related adverse events (AEs) in people with RA, autoimmune diseases (AIDs; rheumatic [r] and non-rheumatic [nr]) and healthy controls (HCs). The survey was circulated by the COVAD study group, comprising 157 collaborators across 106 countries, from February to June 2022. Delayed AEs among RA were compared with other rAIDs, nrAIDs and HCs using multivariable binary regression. A total of 7203 participants were included (1423 [19.7%] RA, 2620 [36.4%] rAIDs, 426 [5.9%] nrAIDs, 2734 [38%] HCs), with 75% female. Compared to HCs, individuals with RA reported higher overall major AEs [OR 1.3 (1.0-1.7)], and an increased number of several minor AEs. Compared to nrAIDs, people with RA had several increased reported minor AEs including myalgia and joint pain. People with active RA had increased major AEs [OR 1.8 (1.1-3.0)] and hospitalisation [OR 4.1 (1.3 - 13.3)] compared to inactive RA. RA patients without autoimmune comorbidities had significantly fewer major and minor AEs than those with other rAIDs. A decreased incidence of hospitalisation was seen in patients taking methotrexate or TNF inhibitors compared to patients not taking these medications. COVID-19 vaccination is associated with minimal to no risks of delayed AEs in patients with RA compared to HCs, and fewer compared to other rAIDs. Active RA and presence of co-existing rAIDs were associated with an increased risk of delayed AEs.
Collapse
Affiliation(s)
- Mrinalini Dey
- Centre for Rheumatic Diseases, King's College London, London, UK
| | - Bohdana Doskaliuk
- Department of Pathophysiology, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Chris Wincup
- Rheumatology Department, King's College Hospital, London, UK
| | - Mrudula Joshi
- Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India
| | - Dzifa Dey
- Department of Medicine and Therapeutics, Rheumatology Unit, University of Ghana Medical School, College of Health Sciences, Korle-Bu, Accra, Ghana
| | - Wanruchada Katchamart
- Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Esha Kadam
- Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India
| | - Parikshit Sen
- Maulana Azad Medical College, 2-Bahadurshah Zafar Marg, New Delhi, Delhi, 110002, India
| | - Samuel Katsuyuki Shinjo
- Division of Rheumatology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Arvind Nune
- Southport and Ormskirk Hospital NHS Trust, Southport, PR8 6PN, UK
| | | | - Nelly Ziade
- Rheumatology Department, Saint-Joseph University, Beirut, Lebanon
- Rheumatology Department, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Yi Ming Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Lisa S Traboco
- Department of Medicine, Section of Rheumatology, St. Luke's Medical Center-Global City, Taguig, Philippines
| | - Carlos Enrique Toro Gutiérrez
- Reference Center for Osteoporosis, Rheumatology and Dermatology, Pontifica Universidad Javeriana Cali, Cali, Colombia
| | - Binit Vaidya
- Department of Rheumatology, National Centre for Rheumatic Diseases, Ratopul, Kathmandu, Nepal
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Latika Gupta
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Centre for Musculoskeletal ResearchManchester Academic Health Science CentreThe University of Manchester, Manchester, UK
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King's College London, London, UK.
- Rheumatology Department, King's College Hospital, London, UK.
| |
Collapse
|
174
|
Tangkum P, Kasitanon N, Gumtorntip W, Winichakoon P, Konsamun S, Wongthanee A, Louthrenoo W. COVID-19 Vaccination in Patients With Systemic Lupus Erythematosus: Adverse Events and Rating Agreement of Flares Between Patients and Physicians. Int J Rheum Dis 2024; 27:e70001. [PMID: 39655469 DOI: 10.1111/1756-185x.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVES To compare adverse events and flares among different doses and types of COVID-19 vaccines in patients with systemic lupus erythematosus (SLE). METHODS All consecutive SLE patients in a lupus cohort, seen between March and October 2022, were invited to join this retrospective study. Inclusion criteria were aged ≥ 20 years and had received at least one dose of COVID-19 vaccine. Data regarding adverse events after vaccination, clinical disease activity and flares within 30 days postvaccination were reviewed. RESULTS A total of 201 SLE patients received 524 vaccine doses, with 201, 199, and 124 patients received 1, 2, and 3 doses, respectively. The vaccines included inactivated virus vaccine, adenovirus-vectored vaccine, and mRNA vaccines in 183 (35%), 128 (24%), and 213 (41%) doses, respectively. Regardless of the dose and type of vaccine, adverse events occurred in 50%-70% of patients. Pain and swelling at the injection site were common local symptoms, whereas constitutional, neurological, musculoskeletal, and mucocutaneous symptoms were among systemic ones. The majority of these symptoms were mild to moderate. Patients reported they had disease flares after vaccination in 5%-6%, while actual flares determined by physicians occurred in 8%-13% of them, giving fair to moderate rating agreement between patients and physicians (Cohen's kappa: 0.21-0.44). There was no significant difference in mean mSLEDAI-2K between pre- and 30 days postvaccination. CONCLUSIONS Adverse events after vaccination were common, regardless of the dose or type of COVID-19 vaccines, but only a small proportion of patients had severe symptoms. Flares were uncommon. The rating agreement of flares between patients and physicians as fair to moderate.
Collapse
Affiliation(s)
- Punsita Tangkum
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nuntana Kasitanon
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wanitcha Gumtorntip
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Poramed Winichakoon
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Supparat Konsamun
- Research Unit, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Antika Wongthanee
- Research Unit, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Worawit Louthrenoo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
175
|
Kadam E, Javaid M, Sen P, Saha S, Ziade N, Day J, Wincup C, Andreoli L, Parodis I, Tan AL, Shinjo SK, Dey D, Cavagna L, Chatterjee T, Knitza J, Wang G, Dalbeth N, Velikova T, Battista S, Cheng K, Boyd P, Kobert L, Gracia-Ramos AE, Mittal S, Makol A, Gutiérrez CET, Caballero-Uribe CV, Kuwana M, Burmester GR, Guillemin F, Nikiphorou E, Chinoy H, Agarwal V, Gupta L. Correction to: Collating the voice of people with autoimmune diseases: Methodology for the third phase of the COVAD studies. Rheumatol Int 2024; 44:3157-3162. [PMID: 39400562 PMCID: PMC11618184 DOI: 10.1007/s00296-024-05692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Esha Kadam
- Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India
| | | | - Parikshit Sen
- Maulana Azad Medical College, 2-Bahadurshah Zafar Marg, New Delhi, Delhi, 110002, India
| | - Sreoshy Saha
- Mymensingh Medical College, Mymensingh, Bangladesh
| | - Nelly Ziade
- Rheumatology Department, Saint-Joseph University, Beirut, Lebanon
- Rheumatology Department, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Jessica Day
- Department of Rheumatology, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Chris Wincup
- Department of Rheumatology, King's College Hospital, London, UK
| | - Laura Andreoli
- Rheumatology and Clinical Immunology Unit, ASST Spedali, Civili and University of Brescia, 25123, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, 25123, Brescia, Italy
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ai Lyn Tan
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Samuel Katsuyuki Shinjo
- Division of Rheumatology, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Dzifa Dey
- Department of Medicine and Therapeutics, School of Medicine and Dentistry, University of Ghana, College of Health Sciences, Korle-Bu, Accra, Ghana
| | - Lorenzo Cavagna
- Rheumatology Unit, Dipartimento Di Medicine Interna E Terapia Medica, Università Degli Studi Di Pavia, Pavia, Lombardy, Italy
| | - Tulika Chatterjee
- Department of Internal Medicine, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Johannes Knitza
- Medizinische Klinik 3, Rheumatologie Und Immunologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Deutschland
| | - Guochun Wang
- Department of Rheumatology, China-Japan Friendship Hospital, Yinghua East Road, Chaoyang District, Beijing, 100029, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University, St. Kliment Ohridski, 1 Kozyak Str., 1407, Sofia, Bulgaria
| | - Simone Battista
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Campus of Savona, University of Genova, Savona, Italy
| | - Karen Cheng
- Associate Board Member, International Myositis Society, Göttingen, Germany
- Patient Advisor, MIHRA Foundation, New Orleans, USA
- Patient Research Partner, Myositis Support and Understanding, Lincoln, USA
| | - Peter Boyd
- Services Support Ofcer, Arthritis Ireland, Dublin, Ireland
- EULAR PARE Committee, Kilchberg, Switzerland
| | - Linda Kobert
- Research and Communications Specialist, The Myositis Association, Columbia, MD, USA
| | - Abraham Edgar Gracia-Ramos
- Department of Internal Medicine, National Medical Center "La Raza", Instituto Mexicano del Seguro Social, General Hospital, Av. Jacaranda S/N, Col. La Raza, Del. Azcapotzalco, C.P. 02990, Mexico City, Mexico
| | - Srijan Mittal
- Maulana Azad Medical College, 2-Bahadurshah Zafar Marg, New Delhi, Delhi, 110002, India
| | - Ashima Makol
- Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Carlos Enrique Toro Gutiérrez
- Reference Center for Osteoporosis, Rheumatology and Dermatology, Pontifcia Universidad Javeriana Cali, Cali, Colombia
| | | | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-Ku, Tokyo, 113-8602, Japan
| | - Gerd-Rüdiger Burmester
- Department of Rheumatology and Clinical Immunology, Charité, University Medicine Berlin, Free University and Humboldt University Berlin, Berlin, Germany
| | - Francis Guillemin
- EA 4360 Apemac, Université de Lorraine, Nancy, France
- Inserm, CHRU Nancy, CIC-1433 Epidémiologie Clinique, Université de Lorraine, Nancy, France
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King's College London, London, UK
- Rheumatology Department, King's College Hospital, London, UK
| | - Hector Chinoy
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, UK
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, UK
- Department of Rheumatology, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Latika Gupta
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, UK.
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK.
| |
Collapse
|
176
|
Balaji D, Balakrishnan R, Srinivasan D, Subbarayan R, Shrestha R, Srivastava N, Chauhan A. The Impact of SARS-CoV-2 on Liver Diseases and Potential Phytochemical Treatments. INFECTIOUS MICROBES AND DISEASES 2024; 6:177-188. [DOI: 10.1097/im9.0000000000000161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has brought about numerous challenges. One of these challenges is the impact of SARS-CoV-2 on the liver. Although this virus primarily affects the lungs, it can induce elevated transaminase levels and the development of scar tissue in the liver, exacerbating preexisting liver conditions. Individuals with preexisting conditions, such as nonalcoholic fatty liver disease, alcohol-induced liver disease and hepatocellular carcinoma, face an increased risk of mortality from COVID-19. However, drugs currently used to treat COVID-19 have undesirable side effects, which make them unsuitable for patients with preexisting liver conditions. In this review, we explore the potential of phytochemicals, such as apigenin, berberine, curcumin, epigallocatechin-3-gallate, quercetin, resveratrol and silymarin, for treatment of the liver conditions, including nonalcoholic fatty liver disease, alcohol-induced liver disease and hepatocellular carcinoma. We also discuss significant associations between phytochemicals and COVID-19 by depicting their molecular interactions. Based on the discussed overlapping functions, it is important to assess the therapeutic efficacy of phytochemicals that possess hepatoprotective properties as potential alternative treatments for COVID-19.
Collapse
Affiliation(s)
- Dhanvee Balaji
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | | | | | | | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
177
|
Kaibagarova I, Saparbaev S, Aringazina R, Zhumabaev M, Nurgaliyeva Z. The role of fetal pancreatic islet cell transplantation in the treatment of type 2 diabetes mellitus. J Diabetes Metab Disord 2024; 23:1949-1957. [PMID: 39610528 PMCID: PMC11599508 DOI: 10.1007/s40200-024-01448-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/24/2024] [Indexed: 11/30/2024]
Abstract
Objectives Diabetes mellitus has a negative impact on patients' lives and is a significant medical and social problem. Due to the high prevalence of diabetes mellitus, shortage of donor materials, immune rejection of the pancreas and limited efficacy of existing treatment methods, the study of promising and more effective approaches to the treatment of this disease, such as transplantation of fetal pancreatic islet cells, becomes relevant. The aim of the study is to determine the efficacy and necessity of fetal pancreatic islet cell transplantation in the treatment of type 2 diabetes mellitus. Methods The study was carried out with the help of analytical-synthetic method, literature review and analysis of medical databases corresponding to the topic of work, clinical and experimental studies conducted by other authors were considered. Results As a result of this work, it was found that the use of fetal stem cell transplantation is an effective method in the treatment of diabetes. Studies confirm that this method reduces hyperglycaemia and NOMA index, increases c-peptide values without serious side effects on the background of treatment. Conclusions Fetal islet cells have advantages in cell culture, relatively low immunogenicity, effective engraftment, although they may produce less insulin relative to adult somatic stem cells. Transplanted islet cells are able to replace and renew the function of the recipient's own pancreatic β-cells, and prevent their destruction. Fetal pancreatic islet cell transplantation is a promising treatment option for type 2 diabetes that can complement or replace existing therapies, improving patients' glucose control.
Collapse
Affiliation(s)
- Indira Kaibagarova
- Department of Pharmacology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Str, Aktobe, 030012 Republic of Kazakhstan
| | - Samat Saparbaev
- Medical Center Al-Jami, 23 Mailin Str, Astana, 010000 Republic of Kazakhstan
| | - Raisa Aringazina
- Department of Internal Diseases No. 1, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Str., 030012 Aktobe, Republic of Kazakhstan
| | - Marat Zhumabaev
- Department of Surgical Diseases No. 1, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Str., 030012 Aktobe, Republic of Kazakhstan
| | - Zhansulu Nurgaliyeva
- Department of Pharmacology, West Kazakhstan Marat Ospanov Medical University, 68 Maresyev Str, Aktobe, 030012 Republic of Kazakhstan
| |
Collapse
|
178
|
Dhanasekaran P, Karasu BT, Mak A. Safety, efficacy, and immunogenicity of SARS-CoV-2 mRNA vaccination in children and adult patients with rheumatic diseases: a comprehensive literature review. Rheumatol Int 2024; 44:2757-2794. [PMID: 39576327 DOI: 10.1007/s00296-024-05734-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/01/2024] [Indexed: 01/03/2025]
Abstract
Patients with autoimmune inflammatory rheumatic diseases (AIIRD) are potentially at a higher risk of contracting the SARS-CoV-2 virus and have poorer outcomes of the infection as a result of their immunocompromised state due to the nature of the underlying autoimmune conditions and immunosuppressant use. mRNA-based vaccines provide a novel approach to establishing immunity against SARS-CoV-2. However, the implications of toll-like receptors (TLRs), type I interferon (IFN) and pro-inflammatory cytokines raise concerns on disease severity and inefficient immune response following mRNA vaccination. The use of immunosuppression to reduce disease activity may have consequential implications on immune responses following SARS-CoV-2 mRNA vaccination. This study systematically reviews the literature on the safety, efficacy, and immunogenicity of SARS-CoV-2 vaccination in patients with autoimmune rheumatic conditions. This comprehensive review was conducted in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). A comprehensive literature search on "PubMed" and "EMBASE" electronic databases was conducted to identify relevant articles published from January 1, 2020 to August 31, 2023. The search yielded 106 studies. The mRNA-based vaccines were demonstrated to be safe and efficacious in AIIRD patients. Most studies investigating safety and efficacy of the mRNA-based vaccines reported low frequencies of serious adverse events and disease flares and few breakthrough infections after complete vaccination. Immunogenic response, however, appeared to be blunted in this population of patients, particularly in those who received certain immunosuppressive agents such as methotrexate, mycophenolic acid and rituximab. mRNA-based vaccines are generally safe and efficacious and produce adequate humoral response in AIIRD patients. Additional prospective studies are warranted to ascertain the long-term safety and efficacy profile and the duration of mRNA-vaccine induced immune response. This can aid in shaping guidelines surrounding optimal timing for booster doses in AIIRD patients.
Collapse
Affiliation(s)
- Preeti Dhanasekaran
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Biraveena Thirunavuc Karasu
- Division of Rheumatology, Department of Medicine, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Rheumatology, Department of Medicine, University Medicine Cluster, National University Health System, Singapore, Singapore
| |
Collapse
|
179
|
Yan J, Yang L, Ren Q, Zhu C, Du H, Wang Z, Qi Y, Xian X, Chen D. Gut microbiota as a biomarker and modulator of anti-tumor immunotherapy outcomes. Front Immunol 2024; 15:1471273. [PMID: 39669573 PMCID: PMC11634861 DOI: 10.3389/fimmu.2024.1471273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/25/2024] [Indexed: 12/14/2024] Open
Abstract
Although immune-checkpoint inhibitors (ICIs) have significantly improved cancer treatment, their effectiveness is limited by primary or acquired resistance in many patients. The gut microbiota, through its production of metabolites and regulation of immune cell functions, plays a vital role in maintaining immune balance and influencing the response to cancer immunotherapies. This review highlights evidence linking specific gut microbial characteristics to increased therapeutic efficacy in a variety of cancers, such as gastrointestinal cancers, melanoma, lung cancer, urinary system cancers, and reproductive system cancers, suggesting the gut microbiota's potential as a predictive biomarker for ICI responsiveness. It also explores the possibility of enhancing ICI effectiveness through fecal microbiota transplantation, probiotics, prebiotics, synbiotics, postbiotics, and dietary modifications. Moreover, the review underscores the need for extensive randomized controlled trials to confirm the gut microbiota's predictive value and to establish guidelines for microbiota-targeted interventions in immunotherapy. In summary, the article suggests that a balanced gut microbiota is key to maximizing immunotherapy benefits and calls for further research to optimize microbiota modulation strategies for cancer treatment. It advocates for a deeper comprehension of the complex interactions between gut microbiota, host immunity, and cancer therapy, aiming for more personalized and effective treatment options.
Collapse
Affiliation(s)
- Jiexi Yan
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lu Yang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhu
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Haiyun Du
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhouyu Wang
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| | - Yaya Qi
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaohong Xian
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Dongsheng Chen
- The State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, Jiangsu, China
| |
Collapse
|
180
|
Criscuolo E, Giuliani B, Castelli M, Cavallaro M, Sisti S, Burioni R, Ferrari D, Mancini N, Locatelli M, Clementi N. Single spike mutation differentiating XBB.1 and XBB.1.5 enhances SARS-CoV-2 cell-to-cell transmission and facilitates serum-mediated enhancement. Front Immunol 2024; 15:1501200. [PMID: 39664381 PMCID: PMC11631925 DOI: 10.3389/fimmu.2024.1501200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction The ongoing emergence of SARS-CoV-2 variants poses significant challenges to existing therapeutics. The spike (S) glycoprotein is central to both viral entry and cell-to-cell transmission via syncytia formation, a process that confers resistance to neutralizing antibodies. The mechanisms underlying this resistance, particularly in relation to spike-mediated fusion, remain poorly understood. Methods We analyzed two clinical SARS-CoV-2 isolates differing by a single amino acid substitution in the S protein. Using biochemical and cell-based assays, we evaluated entry kinetics, syncytia formation, and the neutralizing efficacy of convalescent sera. These parameters were further correlated with S-mediated cell-cell fusion activity. Results The single amino acid substitution significantly altered entry kinetics and enhanced syncytia formation. This modification did not diminished the neutralizing capacity of convalescent sera, but it increased the efficiency of S-induced cell-cell fusion. These findings highlight the mutation's impact on viral transmissibility and immune evasion. Discussion Our study demonstrates that even minor changes in the S protein can profoundly influence SARS-CoV-2 transmissibility and resistance to antibody-mediated neutralization. Understanding the molecular basis of S-mediated cell-cell fusion is crucial for anticipating the impact of emerging variants and developing next-generation therapeutic strategies. These insights provide a framework for predicting variant fitness and optimizing treatment approaches against future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Elena Criscuolo
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Benedetta Giuliani
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Castelli
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Cavallaro
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Sofia Sisti
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | - Roberto Burioni
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
| | | | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, Fondazione Macchi University Hospital, Varese, Italy
| | | | - Nicola Clementi
- Laboratory of Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
181
|
Bartha-Tatár A, Sinkovits G, Schnur J, Maráczi V, Dávid M, Zsigmond B, Rimanóczy É, Szalay B, Biró E, Bekő G, Varga P, Szabó T, Fagyas M, Fejes Z, Kappelmayer J, Nagy B. Prognostic Value of Baseline Serum Pro-Inflammatory Cytokines in Severe Multisystem Inflammatory Syndrome in Children. J Clin Med 2024; 13:7177. [PMID: 39685637 DOI: 10.3390/jcm13237177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Severe clinical manifestations of multisystem inflammatory syndrome in children (MIS-C) are associated with the dysregulation of immune response following SARS-CoV-2 infection. Therefore, we analyzed the levels of 10 selected cytokines at admission to estimate disease severity and to predict the length of hospitalization. In remission samples, these mediators were followed after intravenous immunoglobulin (IVIG) treatment before discharge. Methods: Thirty-five MIS-C patients at the age of 8.4 ± 4.1 years and 11 clinical controls were included. Acute MIS-C patients were divided into two severity subgroups based on their clinical score determined by the WHO criteria. Serum concentrations of IFN-γ, IL-1α, IL-1RA, IL-8, IL-10, IL-17A, IL-18, IP-10, MCP-1, and TNF-α were measured by MILLIPLEX® Human Cytokine/Chemokine panel, while ACE2 activity was determined by a fluorescent kinetic assay. These results were correlated with routinely determined laboratory parameters and clinical characteristics. Results: MIS-C patients demonstrated significantly elevated baseline levels of most of these cytokines compared to controls. Even higher concentrations of IL-18, TNF-α and ferritin with reduced lymphocyte count were found in severe subjects with elevated clinical scores of 4-5 compared to moderate cases with a clinical score of 1-3. Furthermore, the development of cardiovascular dysfunction and prolonged hospitalization (≥8 days) were related to augmented ACE2 and IL-6 levels. IL-18, IL-1RA, IL-10 and TNF-α were diminished in response to IVIG treatment in remission samples. Finally, pre-treatment IL-18 (≥516.8 pg/mL) and TNF-α (≥74.2 pg/mL) effectively differentiated disease severity in MIS-C with AUC values of 0.770 and 0.750, respectively. Conclusions: IL-18 and TNF-α have a prognostic value in disease severity at admission and are capable of monitoring the efficacy of IVIG treatment in MIS-C.
Collapse
Affiliation(s)
- Anita Bartha-Tatár
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Kálmán Laki, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - György Sinkovits
- Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - János Schnur
- Heim Pál National Pediatric Institute, 1089 Budapest, Hungary
| | | | - Máté Dávid
- Heim Pál National Pediatric Institute, 1089 Budapest, Hungary
| | | | - Éva Rimanóczy
- Heim Pál National Pediatric Institute, 1089 Budapest, Hungary
| | - Balázs Szalay
- National Institute of Hematology and Infectious Disease, 1097 Budapest, Hungary
| | - Edina Biró
- National Institute of Hematology and Infectious Disease, 1097 Budapest, Hungary
| | - Gabriella Bekő
- National Institute of Hematology and Infectious Disease, 1097 Budapest, Hungary
| | - Petra Varga
- Institute of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamás Szabó
- Institute of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Miklós Fagyas
- Department of Cardiology, Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
182
|
Pan H, Meng R, Jia Z, Zhang J, Ma W, Liu Y, Wang Q, Li Q. Exercise: a non-drug strategy of NK cell activation. Braz J Med Biol Res 2024; 57:e14144. [PMID: 39607207 DOI: 10.1590/1414-431x2024e14144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
Natural killer (NK) cells are a critical component of the innate immune system and one of the immune cells most sensitive to exercise. So far, it is widely believed that moderate exercise can significantly enhance the proliferation and activity of NK cells, strengthening immune function. However, the impact of exercise on NK cells is a dynamic and complex process. In addition to the type of exercise, the frequency, intensity, and duration of exercise are also key factors. This article not only briefly summarizes the activation mechanisms of NK cells but also delves into the potential importance of exercise as a non-pharmacological strategy in modulating NK cell activity and enhancing the immune system. Emerging studies have indicated that the timing and regularity of exercise bouts might also influence NK cell responses. Moreover, the interaction between exercise and other components of the immune system, such as cytokines and chemokines, could further modulate the functionality of NK cells. The above research is of crucial significance for achieving a deeper understanding of the intricate connection between exercise and NK cell function, as well as the development of effective health promotion strategies. In addition, further research is needed to investigate the effects of long-term exercise on NK cell function and the interaction between exercise and NK cell-mediated immune responses. Translating these research findings into precisely tailored exercise programs for specific populations, taking into account factors like age, health status, and genetic predisposition, could potentially offer unprecedented prospects for further advancements in this burgeoning field of study.
Collapse
Affiliation(s)
- Huixin Pan
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Rui Meng
- School of Data Science, The Chinese University of Hong Kong, Shenzhen, China
| | - Zixuan Jia
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Jing Zhang
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Wen Ma
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Youhan Liu
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Qinglu Wang
- College of Sport and Health, Shandong Sport University, Jinan, China
| | - Qiaoqiao Li
- Key Laboratory of Biomedical Engineering & Technology of Shandong High School, Qilu Medical University, Zibo, China
| |
Collapse
|
183
|
Fu T, Mohan M, Bose M, Brennan EP, Kiriazis H, Deo M, Nowell CJ, Godson C, Cooper ME, Zhao P, Kemp-Harper BK, Woodman OL, Ritchie RH, Kantharidis P, Qin CX. Lipoxin A 4 improves cardiac remodeling and function in diabetes-associated cardiac dysfunction. Cardiovasc Diabetol 2024; 23:413. [PMID: 39563316 PMCID: PMC11577589 DOI: 10.1186/s12933-024-02501-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Diabetic heart disease may eventually lead to heart failure, a leading cause of mortality in diabetic individuals. The lack of effective treatments for diabetes-induced heart failure may result from a failure to address the underlying pathological processes, including chronic, low-grade inflammation. Previous studies have reported that lipoxin A4 (LXA4), known to promote resolution of inflammation, attenuates diabetes-induced atherosclerosis, but its impact on diabetic hearts has not been sought. Thus, we aimed to determine whether LXA4 therapeutic treatment attenuates diabetes-induced cardiac pathology. METHODS Six-week-old male apolipoprotein E-deficient (ApoE-/-) mice were followed for 16 weeks after injection of streptozotocin (STZ, 55 mg/kg/day, i.p. for 5 days) to induce type-1 diabetes (T1DM). Treatment with LXA4 (5 μg/kg, i.p.) or vehicle (0.02% ethanol, i.p.) was administered twice weekly for the final 6 weeks. One week before endpoint, echocardiography was performed within a subset of mice from each group. At the end of the study, mice were euthanized with sodium pentobarbital (100 mg/kg i.p.) and hearts were collected for ex vivo analysis, including histological assessment, gene expression profiling by real-time PCR and protein level measurement by western blot. RESULTS As expected diabetic mice showed a significant elevation in plasma glycated hemoglobin (HbA1c) and glucose levels, along with reduced body weight. Vehicle-treated diabetic mice exhibited increased cardiac inflammation, macrophage content, and an elevated ratio of M1-like to M2-like macrophage markers. In addition, myocardial fibrosis, cardiomyocytes apoptosis and hypertrophy (at the genetic level) were evident, with echocardiography revealing early signs of left ventricular (LV) diastolic dysfunction. Treatment with LXA4 ameliorated diabetes-induced cardiac inflammation, pro-inflammatory macrophage polarization and cardiac remodeling (especially myocardial fibrosis and cardiomyocytes apoptosis), with ultimate improvement in cardiac function. Of note, this improvement was independent of glucose control. CONCLUSIONS These findings demonstrated that LXA4 treatment attenuated the extent of cardiac inflammation in diabetic hearts, resulting in limited cardiac remodeling and improved LV diastolic function. This supports further exploration of LXA4-based therapy for the management of diabetic heart disease. The recent development of stable LXA4 mimetics holds potential as a novel strategy to treat cardiac dysfunction in diabetes, paving the way for innovative and more effective therapeutic strategies.
Collapse
MESH Headings
- Animals
- Lipoxins/pharmacology
- Male
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/complications
- Ventricular Remodeling/drug effects
- Diabetic Cardiomyopathies/physiopathology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/drug therapy
- Ventricular Function, Left/drug effects
- Mice, Knockout, ApoE
- Anti-Inflammatory Agents/pharmacology
- Fibrosis
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/physiopathology
- Diabetes Mellitus, Type 1/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/drug therapy
- Ventricular Dysfunction, Left/prevention & control
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocardium/pathology
- Myocardium/metabolism
- Mice
- Apoptosis/drug effects
- Inflammation Mediators/metabolism
Collapse
Affiliation(s)
- Ting Fu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Muthukumar Mohan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Renal Electrolyte and Hypertension Division, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Madhura Bose
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Eoin P Brennan
- Diabetes Complications Research Centre, School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Minh Deo
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Catherine Godson
- Diabetes Complications Research Centre, School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Peishen Zhao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Owen L Woodman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Phillip Kantharidis
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.
| |
Collapse
|
184
|
Garg P, Singhal G, Kulkarni P, Horne D, Salgia R, Singhal SS. Artificial Intelligence-Driven Computational Approaches in the Development of Anticancer Drugs. Cancers (Basel) 2024; 16:3884. [PMID: 39594838 PMCID: PMC11593155 DOI: 10.3390/cancers16223884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been a complex, resource-intensive process, but AI introduces new opportunities to accelerate discovery, reduce costs, and optimize efficiency. This manuscript delves into the transformative applications of AI-driven methodologies in predicting and developing anticancer drugs, critically evaluating their potential to reshape the future of cancer therapeutics while addressing their challenges and limitations.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Gargi Singhal
- Department of Medical Sciences, S.N. Medical College, Agra 282002, Uttar Pradesh, India
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
185
|
Ishibashi C, Nakanishi K, Nishida M, Shinomiya H, Shinzawa M, Kanayama D, Yamamoto R, Kudo T, Nagatomo I, Yamauchi-Takihara K. Myostatin as a plausible biomarker for early stage of sarcopenic obesity. Sci Rep 2024; 14:28629. [PMID: 39562792 PMCID: PMC11577097 DOI: 10.1038/s41598-024-79534-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024] Open
Abstract
Since sarcopenic obesity (SO) impacts negatively on our health, early detection of SO is essential. However, prevalence of SO in an apparently healthy population has not been well examined. This study aimed to elucidate the prevalence and related factors of SO in middle-aged women, and to investigate useful diagnostic criteria for SO. Body component analyses were conducted on 432 female Osaka University employees aged 30-59 during their health checkups. Healthy (H) and SO groups were defined using cutoff values of 5.7 kg/m2 for skeletal muscle mass index and 30% for percent body fat. Serum myostatin and insulin levels were additionally measured. Among 432 participants, the prevalence of SO was 6.3%. Grip strength (P < 0.0001) was lower and triglyceride (P = 0.0004) and low-density lipoprotein cholesterol (P = 0.0105) levels, and Homeostatic Model Assessment of Insulin Resistance (P = 0.0262) were higher in the SO group than in the H group. Serum myostatin levels in the SO group were lower than in the H group (3,107 pg/mL vs. 3,957 pg/mL, P = 0.0003). Myostatin levels may be suppressed in individuals with SO without any pre-existing conditions. Our diagnostic criteria for SO could reveal the risks for metabolic-related diseases and may be useful for the early detection of SO.
Collapse
Affiliation(s)
- Chisaki Ishibashi
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kaori Nakanishi
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.
| | - Makoto Nishida
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Haruki Shinomiya
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Maki Shinzawa
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Daisuke Kanayama
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Ryohei Yamamoto
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Takashi Kudo
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Izumi Nagatomo
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Keiko Yamauchi-Takihara
- Health Care Division, Health and Counseling Center, Osaka University, 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| |
Collapse
|
186
|
Alqatari S, Alqunais AA, Alali SM, Alharbi MA, Hasan M, Al Shubbar MD. VEXAS Syndrome: A Comprehensive Review of Current Therapeutic Strategies and Emerging Treatments. J Clin Med 2024; 13:6970. [PMID: 39598114 PMCID: PMC11594742 DOI: 10.3390/jcm13226970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
VEXAS syndrome is a recently identified autoinflammatory disorder resulting from somatic mutations in the UBA1 gene, leading to a complex spectrum of severe inflammatory and hematologic manifestations. The absence of established treatment guidelines and the variability in clinical presentation make its management particularly challenging. Current therapeutic approaches are often based on limited evidence, and their effectiveness remains inconsistent. This review seeks to consolidate the existing knowledge on therapeutic strategies for VEXAS syndrome, offering a critical evaluation of their efficacy and addressing the gaps in the current literature. As the clinical recognition of VEXAS grows, there is an urgent need to explore more targeted, effective treatments that can address both the inflammatory and hematologic aspects of the disease. By providing a comprehensive analysis of the current therapeutic landscape, this review aims to guide clinicians and researchers toward developing more effective, long-term management strategies for this life-threatening condition.
Collapse
Affiliation(s)
- Safi Alqatari
- Rheumatology Division, Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (S.A.); (M.H.)
| | - Abdulaziz A. Alqunais
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.A.); (S.M.A.)
| | - Shahad M. Alali
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.A.); (S.M.A.)
| | - Mohammed A. Alharbi
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.A.); (S.M.A.)
| | - Manal Hasan
- Rheumatology Division, Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (S.A.); (M.H.)
| | - Mohammed D. Al Shubbar
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.A.); (S.M.A.)
| |
Collapse
|
187
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
188
|
Sedighi S, Faramarzipalangar Z, Mohammadi E, Aghamohammadi V, Bahnemiri MG, Mohammadi K. The effects of curcumin supplementation on inflammatory markers in systemic lupus erythematosus patients: a randomized placebo-controlled trial. Eur J Nutr 2024; 64:8. [PMID: 39546036 DOI: 10.1007/s00394-024-03515-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multisystem involvement. This study was designed to examine the effects of curcumin, a polyphenolic compound isolated from turmeric rhizomes, on inflammatory markers in SLE patients. METHODS Seventy 18-60 years old SLE patients were recruited in this randomized triple-blinded placebo-controlled trial, and 62 completed the study. Curcumin group received 1000 mg curcumin daily and the placebo group received placebo capsules for 10 weeks. Dietary intakes and serum levels of complement C3 and C4, complement hemolytic 50%, rheumatoid factor, anti-double stranded DNA (anti-ds DNA), erythrocyte sedimentation rate, high-sensitivity C-reactive protein, interlukine-6 (IL-6) and tumor necrosis factor-α were assessed before and after intervention period. RESULTS Curcumin supplementation caused a significant reduction in anti-ds DNA and IL-6 levels at the end of the trial in comparison with baseline (52.57 ± 40.21 vs. 43.27 ± 28.34, p = 0.014 and 127.11 ± 76.63 vs. 101.49 ± 59.08, p = 0.002, respectively). Analysis of covariance which was adjusted for confounding variables also revealed that anti-ds DNA and IL-6 levels decreased significantly in curcumin group compared to placebo group by the end of the intervention period (change:-9.30 ± 19.59 vs. -2.55 ± 17.55, p = 0.018 and - 25.62 ± 42.33 vs. -7.34 ± 34.32, p = 0.043, respectively). No significant changes were observed in levels of other variables during the study (p > 0.05). CONCLUSION Curcumin as an effective and safe adjuvant therapy, ameliorated the autoimmune activity and inflammation in SLE patients via reducing anti-ds DNA and IL-6 levels. CLINICAL TRIAL REGISTRATION irct.behdasht.gov.ir, identifier: IRCT20210425051077N1.
Collapse
Affiliation(s)
- Sima Sedighi
- Golestan Rheumatology Research Center, Golestan University of Medical Science, Gorgan, Iran
| | - Zeinab Faramarzipalangar
- Department of Nutrition, School of Agricultural Sciences and Veterinary Medicine, University of Padua, Padua, Italy.
| | - Elahe Mohammadi
- Department of Nutrition, Khalkhal University of Medical Sciences, Khalkhal, Iran.
- Department of Nutritional Sciences, School of Health, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | | | - Kamran Mohammadi
- Department of Internal Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
189
|
Dick JK, Sangala JA, Krishna VD, Khaimraj A, Hamel L, Erickson SM, Hicks D, Soigner Y, Covill LE, Johnson AK, Ehrhardt MJ, Ernste K, Brodin P, Koup RA, Khaitan A, Baehr C, Thielen BK, Henzler CM, Skipper C, Miller JS, Bryceson YT, Wu J, John CC, Panoskaltsis-Mortari A, Orioles A, Steiner ME, Cheeran MCJ, Pravetoni M, Hart GT. NK Cell and Monocyte Dysfunction in Multisystem Inflammatory Syndrome in Children. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1452-1466. [PMID: 39392378 PMCID: PMC11533154 DOI: 10.4049/jimmunol.2400395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a severe complication of SARS-CoV-2 infection characterized by multiorgan involvement and inflammation. Testing of cellular function ex vivo to understand the aberrant immune response in MIS-C is limited. Despite strong Ab production in MIS-C, SARS-CoV-2 nucleic acid testing can remain positive for 4-6 wk postinfection. Therefore, we hypothesized that dysfunctional cell-mediated Ab responses downstream of Ab production may be responsible for delayed clearance of viral products in MIS-C. In MIS-C, monocytes were hyperfunctional for phagocytosis and cytokine production, whereas NK cells were hypofunctional for both killing and cytokine production. The decreased NK cell cytotoxicity correlated with an NK exhaustion marker signature and systemic IL-6 levels. Potentially providing a therapeutic option, cellular engagers of CD16 and SARS-CoV-2 proteins were found to rescue NK cell function in vitro. Taken together, our results reveal dysregulation in Ab-mediated cellular responses of myeloid and NK cells that likely contribute to the immune pathology of this disease.
Collapse
Affiliation(s)
- Jenna K. Dick
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | - Jules A. Sangala
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| | | | - Aaron Khaimraj
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Lydia Hamel
- Division of Critical Care, Children’s Hospital and Clinics of Minnesota, Minneapolis, MN
| | - Spencer M. Erickson
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Dustin Hicks
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Yvette Soigner
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Laura E. Covill
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Alexander K. Johnson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Michael J. Ehrhardt
- Division of Bone Marrow Transplantation and Cellular Therapy, Department of Pediatrics, M Health Fairview Masonic Children’s Hospital, Minneapolis, MN
| | - Keenan Ernste
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Petter Brodin
- Unit for Clinical Pediatrics, Department of Women’s and Children’s Health, Karolinska Institute, Solna, Sweden
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| | - Richard A. Koup
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Alka Khaitan
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Carly Baehr
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Beth K. Thielen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | | | - Caleb Skipper
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Jeffrey S. Miller
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Division of Hematology, Oncology, and Transplant, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
| | - Yenan T. Bryceson
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Division of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Broegelmann Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN
| | - Chandy C. John
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Angela Panoskaltsis-Mortari
- Division of Bone Marrow Transplantation and Cellular Therapy, Department of Pediatrics, M Health Fairview Masonic Children’s Hospital, Minneapolis, MN
| | - Alberto Orioles
- Division of Critical Care, Children’s Hospital and Clinics of Minnesota, Minneapolis, MN
| | - Marie E. Steiner
- Divisions of Pediatric Critical Care and Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Maxim C. J. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN
| | - Marco Pravetoni
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA
| | - Geoffrey T. Hart
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN
- Center for Immunology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
190
|
Guo Y, Liu H, Chen B, Zhang K, Meng L, Yan L, Niu Q, Zhang J, Yin G, Li Y. Dysregulated CD38 expression on T cells was associated with rapidly progressive interstitial lung disease in anti-melanoma differentiation-associated gene 5 positive dermatomyositis. Front Immunol 2024; 15:1455944. [PMID: 39588376 PMCID: PMC11586385 DOI: 10.3389/fimmu.2024.1455944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024] Open
Abstract
Background Anti-melanoma differentiation-associated gene 5 positive dermatomyositis (MDA5+ DM) is a life-threatening disease due to rapidly progressive interstitial lung disease (RP-ILD). We aimed to investigate the expression profile of T cell subsets in MDA5+ DM patients, seeking for possible disease-causing T cell subsets and potential biomarkers to distinguish ILD, especially RP-ILD patients. Methods Peripheral blood T cell subpopulations were immunophenotyped in 24 MDA5+ DM patients and 21 healthy controls (HCs) by flow cytometry. The proportion of T cell subsets and clinical characteristics were analyzed. The quantitative determination of cytokines in the plasma was measured by using a microsphere-based immunofluorescence assaying kit. Results The proportion of naïve and CD38+ T cells were much higher, whereas the proportion of central memory T cells were lower in MDA5+ DM patients than in HCs. Notably, the proportion of CD38+CD4+ T cells and CD38+CD8+ T cells on T cells in in RP-ILD group were significantly elevated compared to C-ILD, non-ILD group and HCs. Moreover, serum IFN-α levels were significantly increased in MDA5+ DM patients with RP-ILD. Further, the frequencies of CD38+CD4+ T cells and CD38+CD8+ T cells were positively correlated with IFN-α levels. Finally, ROC analysis indicated that CD38+CD4+ T cells and CD38+CD8+ T cells could be potential biomarkers for predicting ILD/RP-ILD in MDA5+ DM patients. Conclusion Dysregulated CD38 expression on T cell subsets was associated with lung involvement, especially RP-ILD in MDA5+ DM patients. CD38+ T cell subsets could be used as potential biomarkers for predicting ILD/RP-ILD in MDA5+ DM patients.
Collapse
Affiliation(s)
- Yixue Guo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Keyi Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Liye Meng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Qian Niu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| | - Geng Yin
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, China
- Clinical Laboratory Medicine Research Center of West China Hospital, Chengdu, China
| |
Collapse
|
191
|
Veronesi F, Salamanna F, Borsari V, Ruffilli A, Faldini C, Giavaresi G. Unlocking diagnosis of sarcopenia: The role of circulating biomarkers - A clinical systematic review. Mech Ageing Dev 2024; 222:112005. [PMID: 39521148 DOI: 10.1016/j.mad.2024.112005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Sarcopenia, the gradual loss of muscle mass, strength, and function with age, poses a significant risk to older adults, making early diagnosis crucial for preventing disability and enhancing quality of life. Biomarkers are vital for the early detection, monitoring progression, and assessing the efficacy of treatments for sarcopenia, offering a detailed evaluation of muscle health. This systematic review examined the clinical potential of circulating biomarkers in sarcopenia by analyzing studies up to May 2024 from PubMed, Scopus, Web of Science. A total of 45 studies involving 641,730 patients were reviewed, revealing notable biomarker differences between sarcopenic and non-sarcopenic individuals. Sarcopenic patients exhibited lower levels of certain microRNAs, hemoglobin, albumin, and anti-inflammatory factors, alongside higher levels of red and white blood cells, pro-inflammatory factors, growth factors, matrix proteins, free thyroxine, cortisol, and adiponectin. Additionally, they had lower levels of irisin, free triiodothyronine, and insulin, with reduced phosphatidylcholines and elevated spermidine. The studies were generally of fair to good quality, but due to heterogeneity, a meta-analysis was not feasible. The review underscores the need for standardized biomarkers and diagnostic criteria and suggests that improving outcomes for sarcopenic patients may involve addressing inflammation, metabolic, and hormonal issues through nutrition, medication, and exercise.
Collapse
Affiliation(s)
- F Veronesi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy
| | - F Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy.
| | - V Borsari
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy
| | - A Ruffilli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy; Department of Biomedical and Neuromotor Science - DIBINEM, University of Bologna, Bologna, Italy
| | - C Faldini
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy; Department of Biomedical and Neuromotor Science - DIBINEM, University of Bologna, Bologna, Italy
| | - G Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, Bologna 40136, Italy
| |
Collapse
|
192
|
Biennier S, Fontaine M, Duquenoy A, Schwintner C, Doré J, Corvaia N. Narrative Review: Advancing Dysbiosis Treatment in Onco-Hematology with Microbiome-Based Therapeutic Approach. Microorganisms 2024; 12:2256. [PMID: 39597645 PMCID: PMC11596191 DOI: 10.3390/microorganisms12112256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
This review explores the complex relationship between gut dysbiosis and hematological malignancies, focusing on graft-versus-host disease (GvHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. We discuss how alterations in microbial diversity and composition can influence disease development, progression, and treatment outcomes in blood cancers. The mechanisms by which the gut microbiota impacts these conditions are examined, including modulation of immune responses, production of metabolites, and effects on intestinal barrier function. Recent advances in microbiome-based therapies for treating and preventing GvHD are highlighted, with emphasis on full ecosystem standardized donor-derived products. Overall, this review underscores the growing importance of microbiome research in hematology-oncology and its potential to complement existing treatments and improve outcomes for thousands of patients worldwide.
Collapse
Affiliation(s)
- Salomé Biennier
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Aurore Duquenoy
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, 78350 Jouy-en-Josas, France;
| | | |
Collapse
|
193
|
Huang J, Su H, Lin J, Zhu F, Jiang X, Pan J. Achalasia Following SARS-CoV-2 Infection: A Case Report and Review of Literature. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:1943-1947. [PMID: 38657616 DOI: 10.1055/a-2275-2423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
RATIONALE The COVID-19 pandemic caused by the SARS-CoV-2 virus has led to health complications beyond respiratory symptoms, revealing multi-organ involvement, including potential gastrointestinal implications. PATIENT CONCERNS We present a case of a 40-year-old female without any history of achalasia who developed symptoms of the condition following a confirmed SARS-CoV-2 infection. Unusually, multiple esophageal ulcers were identified, which are not typically associated with achalasia. DIAGNOSIS Achalasia and esophageal ulcers were confirmed through a series of examinations, including barium swallow, CT scan, and upper endoscopy. Furthermore, immunohistochemical staining of esophageal biopsy specimens revealed the presence of the SARS-CoV-2 spike protein, suggesting direct viral involvement. INTERVENTIONS The patient was treated with calcium channel blockers and proton pump inhibitors and later underwent a peroral endoscopic myotomy (POEM) procedure following the resolution of her COVID-19 infection. OUTCOME After the POEM procedure, the patient made a good recovery. LESSONS This case underscores the potential for SARS-CoV-2 to trigger gastrointestinal complications and emphasizes the need for ongoing patient management and further research into the long-term implications of COVID-19. Despite the single-case nature of this report, it contributes to the expanding understanding of the diverse and multi-systemic impact of COVID-19.
Collapse
Affiliation(s)
- Jianbin Huang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| | - Huang Su
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| | - Jiejun Lin
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| | - Fangchao Zhu
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| | - Xiaofen Jiang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| | - Jie Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou 325000, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou 325000, Zhejiang province, China
- Department of Gastroenterology, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang province, China
| |
Collapse
|
194
|
Esmailian A, Barnes SL, Pumar M. Case report of secondary T-cell deficiency following the AstraZeneca COVID-19 vaccine. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100339. [PMID: 39380982 PMCID: PMC11459643 DOI: 10.1016/j.jacig.2024.100339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 10/10/2024]
Abstract
We present a case of secondary T-cell deficiency particularly affecting CD4 T cells, along with the emergence of chronic spontaneous urticaria in a patient following COVID-19 vaccination. The condition was partially managed with omalizumab after initial first-line therapy proved ineffective.
Collapse
Affiliation(s)
- Amin Esmailian
- Faculty of Medicine, Monash University, Melbourne, Australia
| | - Sara Laura Barnes
- Faculty of Medicine, Monash University, Melbourne, Australia
- Monash Lung Sleep Allergy and Immunology, Clayton, Australia
- Monash Health, Clayton, Australia
| | | |
Collapse
|
195
|
Babun FK, Kayar NA, Hatipoğlu M. Investigating the role of salivary Interleukin-40 levels in diagnosing periodontal diseases. Oral Dis 2024; 30:5315-5325. [PMID: 38515006 PMCID: PMC11610694 DOI: 10.1111/odi.14936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND The present study aimed to analyze IL-40, IL-1β, and MMP-8 levels in periodontitis as well as gingivitis and periodontal health, and to explore potential correlations between these biomarkers and standard clinical parameters. MATERIALS AND METHODS We collected saliva samples from 120 systemically healthy, non-smoking individuals aged between 18 and 63 years. These individuals were divided into three groups: healthy controls [S], gingivitis [G], and stage III grade B periodontitis [P]. IL-40, IL-1β, and MMP-8 levels in saliva samples were analyzed by ELISA. RESULTS We observed significantly elevated salivary IL-40 levels in the G group compared to the S group (p = 0.003). We found significantly higher salivary IL-1β levels in the P group compared to both the S and G groups (p = 0.000). Salivary MMP-8 levels were significantly higher in the P group than in the S group (p = 0.016). CONCLUSION Our study suggests that IL-40 and IL-1β may serve as effective salivary biomarkers for diagnosing gingivitis, while MMP-8 and IL-1β may be effective for distinguishing periodontitis. Based on our study's findings, it can be stated that IL-40 may serve as a new and effective biomarker for distinguishing individuals with gingivitis from healthy ones.
Collapse
Affiliation(s)
- Fatma Köksel Babun
- Department of PeriodontologyAkdeniz University Faculty of DentistryAntalyaTurkey
| | - Nezahat Arzu Kayar
- Department of PeriodontologyAkdeniz University Faculty of DentistryAntalyaTurkey
| | - Mükerrem Hatipoğlu
- Department of PeriodontologyAkdeniz University Faculty of DentistryAntalyaTurkey
| |
Collapse
|
196
|
Nielipińska D, Rubiak D, Pietrzyk-Brzezińska AJ, Małolepsza J, Błażewska KM, Gendaszewska-Darmach E. Stapled peptides as potential therapeutics for diabetes and other metabolic diseases. Biomed Pharmacother 2024; 180:117496. [PMID: 39362065 DOI: 10.1016/j.biopha.2024.117496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The field of peptide drug research has experienced notable progress, with stapled peptides featuring stabilized α-helical conformation, emerging as a promising field. These peptides offer enhanced stability, cellular permeability, and binding affinity and exhibit potential in the treatment of diabetes and metabolic disorders. Stapled peptides, through the disruption of protein-protein interactions, present varied functionalities encompassing agonism, antagonism, and dual-agonism. This comprehensive review offers insight into the technology of peptide stapling and targeting of crucial molecular pathways associated with glucose metabolism, insulin secretion, and food intake. Additionally, we address the challenges in developing stapled peptides, including concerns pertaining to structural stability, peptide helicity, isomer mixture, and potential side effects.
Collapse
Affiliation(s)
- Dominika Nielipińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| | - Dominika Rubiak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Agnieszka J Pietrzyk-Brzezińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland
| | - Joanna Małolepsza
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland
| | - Katarzyna M Błażewska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Poland.
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Poland.
| |
Collapse
|
197
|
Hoxha A, Striani G, Lovisotto M, Simioni P, Doria A, Ramonda R. COVID-19 vaccine and the risk of flares in inflammatory arthritis: a systematic literature review and meta-analysis. Front Immunol 2024; 15:1503895. [PMID: 39555064 PMCID: PMC11563799 DOI: 10.3389/fimmu.2024.1503895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) vaccines aroused concerns about the risk of flares and adverse events in inflammatory arthritis (IA) since the vaccine clinical trials did not specifically investigate this subset of patients. Methods A systematic literature review and meta-analysis to summarize the data on joint disease flare and adverse events following immunization (AEFI). Two researchers independently evaluated the literature on Pubmed, Scopus, and EMBASE databases from 22nd March 2020 to 30th September 2023. A random-effects model was used to pool odds ratios (OR) (with 95% CI) for the risk of joint disease flares and adverse events. Subgroup analyses were performed to evaluate the risk of disease flare between different IA and adverse events. Heterogeneity was assessed by I2 statistic. Results A total of 9874 IA patients were included in the study: 6579 (66.6%) patients affected by RA and 3295 (33.4%) spondyloarthritis (SpA). The overall rate of flares was higher in RA vs. SpA (9.1% vs. 5.3%). However, the pooled estimated analysis showed no increased risk of joint disease flare following COVID-19 vaccination in patients affected by RA vs. SpA [OR 0.88, 95% CI: 0.77-1.00]. Furthermore, a subgroup analysis showed an increased risk of joint flares in psoriatic arthritis (PsA) patients vs. RA [OR 0.79, 95% CI: 0.68-0.93, p=0.004]. The pooled estimated analysis revealed no increased risk of AEFI in patients with RA vs. SpA [1.02, 95% CI: 0.63-1.65]. Conclusions Our meta-analysis summarized the current evidence on joint disease flares and COVID-19 vaccine-associated AEFI in IA patients. Pooled analysis showed an increased risk of disease flares in PsA vs. RA patients.
Collapse
Affiliation(s)
- Ariela Hoxha
- Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine (DIMED), Internal Medicine Division, Padova University Hospital, Padova, Italy
| | - Giovanni Striani
- Rheumatology Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| | - Marco Lovisotto
- Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine (DIMED), Internal Medicine Division, Padova University Hospital, Padova, Italy
| | - Paolo Simioni
- Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine (DIMED), Internal Medicine Division, Padova University Hospital, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| |
Collapse
|
198
|
Zhao Z, Yang M, Zhao X, Wan F, Ning B, Zhang L, Fu J. The Impact of Food Specific IgG Antibodies on Migraine and Its Comorbidities. Immun Inflamm Dis 2024; 12:e70056. [PMID: 39552191 PMCID: PMC11570760 DOI: 10.1002/iid3.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVE To investigate the differences in headache and comorbidity symptoms between migraine patients with negative and positive food specific IgG antibodies, and explore the correlation between these symptoms with food specific IgG antibodies. METHODS A total of 129 migraine patients were enrolled. Seven questionnaires were used to gather information regarding the symptoms of migraine, gastrointestinal, depression, anxiety, and sleep. Serum specific IgG antibodies against 14 kinds of food were detected using enzyme-linked immunosorbent assays. RESULTS Patients with migraine diagnosis who had positive food specific IgG antibodies had significantly worse headaches, gastrointestinal and anxiety symptoms, compared to the patients with negative IgG antibodies. Patients with more IgG positive foods and higher total positive IgG concentration generally had worse migraine conditions, anxiety, depression, and gastrointestinal symptoms. CONCLUSION The effect of food specific IgG antibodies on severity of migraine and its comorbidities were antibody-quantity and IgG-concentration dependent. Future studies are warranted to explore the mechanism underlying such relationship.
Collapse
Affiliation(s)
- Zhi‐Ming Zhao
- Health Center of Screening and Prevention of DiseasesFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Mei‐Mei Yang
- Department of NeurologyFourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Xian‐Shu Zhao
- Department of GastroenterologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Fu‐Jun Wan
- Health Center of Screening and Prevention of DiseasesFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Bao‐Li Ning
- Health Center of Screening and Prevention of DiseasesFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Li‐Ming Zhang
- Department of NeurologyFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| | - Jun Fu
- Health Center of Screening and Prevention of DiseasesFirst Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangChina
| |
Collapse
|
199
|
Battista S, Giardulli B, Sieiro Santos C, Aharonov O, Puttaswamy D, Russell AM, Gupta L. Digital Health and Self-Management in Idiopathic Inflammatory Myopathies: A Missed Opportunity? Curr Rheumatol Rep 2024; 26:383-391. [PMID: 39115596 PMCID: PMC11469974 DOI: 10.1007/s11926-024-01157-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE OF REVIEW This paper explored the potential of digital health in idiopathic inflammatory myopathies (IIMs), with a focus on self-management. Digital self-management technology includes tailored treatment plans, symptom tracking, educational resources, enhanced communication, and support for long-term planning. RECENT FINDINGS After arguing the importance of digital health in IIMs management, from diagnosis until treatment, our literature review revealed a notable gap in research focusing on the efficacy of digital self-management interventions for individuals with IIMs, with no randomised controlled trials or observational studies addressing this topic. Our review further highlighted the significant unmet need for research in self-management interventions for individuals with IIMs. The absence of studies underscores the necessity for collaborative efforts to address this gap and develop personalised, effective strategies for managing IIMs using digital technology. Individuals with IIMs deserve tailored self-management approaches akin to those available for other rheumatic and musculoskeletal diseases.
Collapse
Affiliation(s)
- Simone Battista
- School of Health and Society, Centre for Human Movement and Rehabilitation, University of Salford, Salford, Greater Manchester, UK
| | - Benedetto Giardulli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Campus of Savona, Savona, Italy
| | | | - Or Aharonov
- Department of Psychology, Psychology of Aging Group, University of Vienna, Vienna, Austria
| | - Darshan Puttaswamy
- Seth Gordhandhas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, Maharashtra, India
| | - Anne -Marie Russell
- School of Medicine and Health, University of Birmingham, Edgbaston, Birmingham, UK
- School of Health and Care Professions, University of Exeter, Exeter, Devon, UK
- ILD Regional Service, University Hospitals Birmingham NHS Trust, Birmingham, UK
| | - Latika Gupta
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK.
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| |
Collapse
|
200
|
Jun HR, Kim YH, Moon JE, Jeong S, Goh HS, Hoang MH, Lee YN, Jeong H, Shim IK, Kim SC. Effect of isoproterenol, a β-adrenergic agonist, on the differentiation of insulin-producing pancreatic β cells derived from human pluripotent stem cells. Exp Cell Res 2024; 443:114307. [PMID: 39461404 DOI: 10.1016/j.yexcr.2024.114307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Research on islet replacement through the differentiation of functionally matured insulin-producing β-like cells for the treatment of diabetes presents a significant challenge. Neural signals in β cell differentiation significantly impact the pancreatic microenvironment in glucose metabolism, but they are not fully understood. In this study, isoproterenol, a β adrenoreceptor agonist, was introduced into pancreatic progenitor cells, derived from human pluripotent stem cells in vitro, undergoing endocrine differentiation, using 2-dimensional (2D) and 3-dimensional (3D) differentiation protocols. This resulted in increased insulin and C-peptide secretion, along with elevated expression of key pancreatic beta cell transcription factors, including PDX-1, NKX6.1, and MAFA, and improved function, demonstrated by increased responsiveness to glucose determined via a glucose-stimulated insulin secretion test. Moreover, RNA transcriptome analysis of isoproterenol-treated endocrine progenitors facilitated the identification of biological pathways and genes that contribute to mature beta cell differentiation efficiency correlated with neural signals, such as adrenoceptor beta 1, calcium/calmodulin dependent protein kinase II alpha, phospholipase C delta 4, and neurotrophic receptor tyrosine kinase 1. Among those genes, calcium/calmodulin dependent protein kinase II alpha was suggested as the most notable gene involved in the isoproterenol mechanism through inhibitor assays. This study illustrates that isoproterenol significantly enhances endocrine differentiation and underscores its effects on stem cell-derived beta cell maturation, emphasizing its therapeutic potential for the treatment of diabetes.
Collapse
Affiliation(s)
- Hye Ryeong Jun
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yang Hee Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Moon
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sehui Jeong
- Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Han Se Goh
- Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minh Hien Hoang
- Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yu Na Lee
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyemin Jeong
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Song Cheol Kim
- Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|