Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24.734
Filter
1.
Front Immunol ; 15: 1380889, 2024.
Article in English | MEDLINE | ID: mdl-38979412

ABSTRACT

Background: Mounting evidence suggests a connection between inflammatory cytokines and adhesive capsulitis (AC). However, the specific systemic inflammatory cytokines contributing to AC have not been clearly identified. This study employed Mendelian randomization (MR) to explore the causal relationships between 41 inflammatory cytokines and AC. Methods: In this bidirectional, two-sample MR analysis, genetic variations associated with AC were derived from a comprehensive genome-wide association study (GWAS). The inflammatory cytokines data were sourced from a GWAS summary involving 8,293 healthy participants. The primary MR method employed was inverse variance weighting, supplemented by MR-Egger, weighted median, and MR-pleiotropy residual sum and outlier for sensitivity analysis. Heterogeneity was assessed using Cochran's Q test, and the MR results were validated using the leave-one-out method. Results: Elevated levels of interferon gamma-induced protein 10 (IP-10) (odds ratio (OR) = 1.086, 95% confidence interval (CI) = 1.002-1.178) and regulated on activation, normal T cell expressed and secreted (RANTES) (OR = 1.107, 95% CI = 1.026-1.195) were linked to an increased risk of AC. Increased levels of stromal cell-derived factor-1 alpha (SDF-1α) (OR = 0.879, 95% CI = 0.793-0.974) and tumor necrosis factor-alpha (TNF-α) (OR = 0.911, 95% CI = 0.831-0.999) were associated with a reduced AC risk. Moreover, genetically predicted AC exhibited associations with elevated cutaneous T cell attracting (CTACK) levels (OR = 1.202, 95% CI = 1.007-1.435) and diminished levels of interleukin-17 (IL-17) (OR = 0.678, 95% CI = 0.518-0.888) and interleukin-5 (IL-5) (OR = 0.786, 95% CI = 0.654-0.944), as confirmed through inverse-variance weighted (IVW) methods. Conclusion: The present study successfully establishes a causal association between genetically proxied circulating levels of IP-10, RANTES, SDF-1α, and TNF-α and the risk of AC. Additionally, AC contributes to an increase in CTACK and a decrease in IL-17 and IL-5. This significant finding not only enhances the understanding of the pathogenesis of AC but also holds promise for the development of effective clinical management strategies.


Subject(s)
Bursitis , Cytokines , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Cytokines/blood , Cytokines/genetics , Bursitis/genetics , Genetic Predisposition to Disease , Inflammation Mediators/metabolism , Inflammation Mediators/blood
2.
Cells ; 13(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38995013

ABSTRACT

Skeletal muscle regeneration after injury is a complex process involving inflammatory signaling and myoblast activation. Pro-inflammatory cytokines like tumor necrosis factor-alpha (TNF-α) are key mediators, but their effects on gene expression in proliferating myoblasts are unclear. We performed the RNA sequencing of TNF-α treated C2C12 myoblasts to elucidate the signaling pathways and gene networks regulated by TNF-α during myoblast proliferation. The TNF-α (10 ng/mL) treatment of C2C12 cells led to 958 differentially expressed genes compared to the controls. Pathway analysis revealed significant regulation of TNF-α signaling, along with the chemokine and IL-17 pathways. Key upregulated genes included cytokines (e.g., IL-6), chemokines (e.g., CCL7), and matrix metalloproteinases (MMPs). TNF-α increased myogenic factor 5 (Myf5) but decreased MyoD protein levels and stimulated the release of MMP-9, MMP-10, and MMP-13. TNF-α also upregulates versican and myostatin mRNA. Overall, our study demonstrates the TNF-α modulation of distinct gene expression patterns and signaling pathways that likely contribute to enhanced myoblast proliferation while suppressing premature differentiation after muscle injury. Elucidating the mechanisms involved in skeletal muscle regeneration can aid in the development of regeneration-enhancing therapeutics.


Subject(s)
Cell Proliferation , Myoblasts , Signal Transduction , Tumor Necrosis Factor-alpha , Myoblasts/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Proliferation/drug effects , Animals , Mice , Cell Line , Chemokines/metabolism , Chemokines/genetics , Cytokines/metabolism , Cytokines/genetics , Gene Expression Regulation/drug effects
3.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38995674

ABSTRACT

Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.


Subject(s)
Alphavirus Infections , Alphavirus , Genotype , Macaca fascicularis , RNA, Viral , Viremia , Animals , Macaca fascicularis/virology , Alphavirus/genetics , Alphavirus/pathogenicity , Alphavirus/classification , Alphavirus/isolation & purification , Alphavirus Infections/virology , Alphavirus Infections/veterinary , Viremia/virology , RNA, Viral/genetics , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Disease Models, Animal , Phylogeny , Cytokines/genetics , Cytokines/blood
4.
Ecotoxicol Environ Saf ; 281: 116664, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38954909

ABSTRACT

BACKGROUND: Observational studies have reported associations between air pollutants and brain imaging-derived phenotypes (IDPs); however, whether this relationship is causal remains uncertain. METHODS: We conducted bidirectional two-sample Mendelian randomization (MR) analyses to explore the causal relationships between 5 types of air pollutants (N=423,796 to 456,380 individuals) and 587 reliable IDPs (N=33,224 individuals). Two-step MR was also conducted to assess whether the identified effects are mediated through the modulation of circulating cytokines (N=8293). RESULTS: We found genetic evidence supporting the association of nitrogen oxides (NOx) with mean intra-cellular volume fraction (ICVF) in the left uncinate fasciculus (IVW ß=-0.42, 95 % CI -0.62 to -0.23, P=1.51×10-5) and mean fractional anisotropy (FA) in the left uncinate fasciculus (IVW ß=-0.42, 95 % CI -0.62 to -0.21, P=4.89×10-5). In further two-step MR analyses, we did not find evidence that genetic predictions of any circulating cytokines mediated the association between NOx and IDPs. CONCLUSION: This study provides evidence for the association between air pollutants and brain IDPs, emphasizing the importance of controlling air pollution to improve brain health.


Subject(s)
Air Pollutants , Air Pollution , Brain , Phenotype , Humans , Air Pollution/adverse effects , Air Pollutants/toxicity , Brain/diagnostic imaging , Mendelian Randomization Analysis , Nitrogen Oxides , Cytokines/genetics , Cytokines/blood , Neuroimaging
5.
Rev Assoc Med Bras (1992) ; 70(7): e20230188, 2024.
Article in English | MEDLINE | ID: mdl-39045924

ABSTRACT

OBJECTIVE: Nonalcoholic fatty liver disease is a chronic liver disease and a growing global epidemic. The aim of this study was to investigate the association between a visfatin gene (NAMPT) variant and nonalcoholic fatty liver disease, owing to the connection between this disease and insulin resistance, obesity, inflammation, and oxidative stress, and the role of visfatin in these metabolic disorders. METHODS: In the present case-control study, we enrolled 312 genetically unrelated individuals, including 154 patients with biopsy-proven nonalcoholic fatty liver disease and 158 controls. The rs2058539 polymorphism of NAMPT gene was genotyped using the PCR-RFLP method. RESULTS: Genotype and allele distributions of NAMPT gene rs2058539 polymorphism conformed to the Hardy-Weinberg equilibrium both in the case and control groups (p>0.05). The distribution of NAMPT rs2058539 genotypes and alleles differed significantly between the cases with nonalcoholic fatty liver disease and controls. The "CC" genotype of the NAMPT rs2058539 compared with "AA" genotype was associated with a 2.5-fold increased risk of nonalcoholic fatty liver disease after adjustment for confounding factors [p=0.034; odds ratio (OR)=2.52, 95% confidence interval (CI)=1.36-4.37]. Moreover, the NAMPT rs2058539 "C" allele was significantly overrepresented in the nonalcoholic fatty liver disease patients than controls (p=0.022; OR=1.77, 95%CI=1.14-2.31). CONCLUSION: Our findings indicated for the first time that the NAMPT rs2058539 "CC" genotype is a marker of increased nonalcoholic fatty liver disease susceptibility; however, it needs to be supported by further investigations in other populations.


Subject(s)
Cytokines , Genetic Predisposition to Disease , Genotype , Nicotinamide Phosphoribosyltransferase , Non-alcoholic Fatty Liver Disease , Polymorphism, Single Nucleotide , Humans , Nicotinamide Phosphoribosyltransferase/genetics , Non-alcoholic Fatty Liver Disease/genetics , Female , Male , Case-Control Studies , Middle Aged , Risk Factors , Adult , Genetic Predisposition to Disease/genetics , Cytokines/genetics , Gene Frequency/genetics , Alleles , Polymorphism, Restriction Fragment Length , Polymerase Chain Reaction
6.
Function (Oxf) ; 5(4)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38985000

ABSTRACT

Pancreatic ß-cells are essential for survival, being the only cell type capable of insulin secretion. While they are believed to be vulnerable to damage by inflammatory cytokines such as interleukin-1 beta (IL-1ß) and interferon-gamma, we have recently identified physiological roles for cytokine signaling in rodent ß-cells that include the stimulation of antiviral and antimicrobial gene expression and the inhibition of viral replication. In this study, we examine cytokine-stimulated changes in gene expression in human islets using single-cell RNA sequencing. Surprisingly, the global responses of human islets to cytokine exposure were remarkably blunted compared to our previous observations in the mouse. The small population of human islet cells that were cytokine responsive exhibited increased expression of IL-1ß-stimulated antiviral guanylate-binding proteins, just like in the mouse. Most human islet cells were not responsive to cytokines, and this lack of responsiveness was associated with high expression of genes encoding ribosomal proteins. We further correlated the expression levels of RPL5 with stress response genes, and when expressed at high levels, RPL5 is predictive of failure to respond to cytokines in all endocrine cells. We postulate that donor causes of death and isolation methodologies may contribute to stress of the islet preparation. Our findings indicate that activation of stress responses in human islets limits cytokine-stimulated gene expression, and we urge caution in the evaluation of studies that have examined cytokine-stimulated gene expression in human islets without evaluation of stress-related gene expression.


Subject(s)
Cytokines , Islets of Langerhans , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Islets of Langerhans/metabolism , Islets of Langerhans/drug effects , Cytokines/metabolism , Cytokines/genetics , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Sequence Analysis, RNA , Stress, Physiological/drug effects , Interleukin-1beta/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Male , Mice , Animals , RNA-Seq , Female , Middle Aged , Single-Cell Gene Expression Analysis
7.
J Am Heart Assoc ; 13(14): e034764, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38979813

ABSTRACT

BACKGROUND: Long noncoding RNA (lncRNA) and mRNA profiles in leukocytes have shown potential as biomarkers for acute ischemic stroke (AIS). This study aimed to identify altered lncRNA and target mRNA profiles in peripheral blood leukocytes as biomarkers and to assess the diagnostic value and association with AIS prognosis. METHODS AND RESULTS: Differentially expressed lncRNAs (DElncRNAs) and differentially expressed target mRNAs (DEmRNAs) were screened by RNA sequencing in the discovery set, which consisted of 10 patients with AIS and 20 controls. Validation sets consisted of a multicenter (311 AIS versus 303 controls) and a nested case-control study (351 AIS versus 352 controls). The discriminative value of DElncRNAs and DEmRNAs added to the traditional risk factors was estimated with the area under the curve. NAMPT-AS, FARP1-AS1, FTH1, and NAMPT were identified in the multicenter case-control study (P<0.05). LncRNA NAMPT-AS was associated with cis-target mRNA NAMPT and trans-target mRNA FTH1 in all validation sets (P<0.001). Similarly, AIS cases exhibited upregulated lncRNA FARP-AS1 and FTH1 expression (P<0.001) in the nested case-control study (P<0.001). Furthermore, lncRNA FARP1-AS1 expression was upregulated in AIS patients at discharge with an unfavorable outcome (P<0.001). Positive correlations were found between NAMPT expression level and NIHSS scores of AIS patients (P<0.05). Adding 2 lncRNAs and 2 target mRNAs to the traditional risk factor model improved area under the curve by 22.8% and 5.2% in the multicenter and the nested case-control studies, respectively. CONCLUSIONS: lncRNA NAMPT-AS and FARP1-AS1 have potential as diagnostic biomarkers for AIS and exhibit good performance when combined with target mRNA NAMPT and FTH1.


Subject(s)
Biomarkers , Ischemic Stroke , Leukocytes , RNA, Long Noncoding , RNA, Messenger , Humans , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Male , Female , Ischemic Stroke/genetics , Ischemic Stroke/diagnosis , Ischemic Stroke/blood , RNA, Messenger/blood , RNA, Messenger/genetics , Middle Aged , Case-Control Studies , Prognosis , Leukocytes/metabolism , Aged , Biomarkers/blood , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/blood , Cytokines/blood , Cytokines/genetics , Reproducibility of Results
8.
PeerJ ; 12: e17477, 2024.
Article in English | MEDLINE | ID: mdl-39006008

ABSTRACT

Objective: The primary objective is to study the impact of gut microbiota and their interactions with diverse immunological markers on the development of rheumatoid arthritis. Methods: This study was performed in Astana, Kazakhstan, and included 77 Kazakh female patients older than 18 years, who met the American College of Rheumatology 2010 classification criteria for rheumatoid arthritis (RA), and 113 healthy controls. The DNA was extracted from fecal samples obtained from all study participants for subsequent sequencing at the 16S rRNA gene V1-V3 locus, facilitating the analysis of the gut microbiome. The Multiplex immunoassay was employed to measure the concentrations of inflammatory cytokines, chemokines, and immunoglobulins in both fecal and plasma samples. Results: Our taxonomic analysis revealed significant differences in the composition of the gut microbiota between the healthy control cohort and the cohort with rheumatoid arthritis RA. Alpha diversity was significantly lower in the RA group. Lachnospiraceae were the most abundant taxon and found to be crucial, showing correlations with immunological markers such as IL5. Additionally, Lachnospiraceae and Oscillospiraceae exhibited the most predictable power and distinguished the composition of both study groups. Conclusion: Our study identifies key differences in the gut microbiome of RA patients, revealing distinct microbial patterns and specific taxa abundance. We highlight potential biomarkers in immunological and bacterial pathways, offering insights into RA development and indicating possibilities for personalized treatment.


Subject(s)
Arthritis, Rheumatoid , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/microbiology , Gastrointestinal Microbiome/immunology , Female , Middle Aged , Adult , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Case-Control Studies , Kazakhstan , Biomarkers/blood , Cytokines/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/blood
9.
Physiol Rep ; 12(13): e16129, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38955668

ABSTRACT

Cardiotrophin-1 (CT-1), a member of the interleukin (IL)-6 cytokine family, has renoprotective effects in mouse models of acute kidney disease and tubulointerstitial fibrosis, but its role in glomerular disease is unknown. To address this, we used the mouse model of nephrotoxic nephritis to test the hypothesis that CT-1 also has a protective role in immune-mediated glomerular disease. Using immunohistochemistry and analysis of single-cell RNA-sequencing data of isolated glomeruli, we demonstrate that CT-1 is expressed in the glomerulus in male mice, predominantly in parietal epithelial cells and is downregulated in mice with nephrotoxic nephritis. Furthermore, analysis of data from patients revealed that human glomerular disease is also associated with reduced glomerular CT-1 transcript levels. In male mice with nephrotoxic nephritis and established proteinuria, administration of CT-1 resulted in reduced albuminuria, prevented podocyte loss, and sustained plasma creatinine, compared with mice administered saline. CT-1 treatment also reduced fibrosis in the kidney cortex, peri-glomerular macrophage accumulation and the kidney levels of the pro-inflammatory mediator complement component 5a. In conclusion, CT-1 intervention therapy delays the progression of glomerular disease in mice by preserving kidney function and inhibiting renal inflammation and fibrosis.


Subject(s)
Cytokines , Kidney Glomerulus , Mice, Inbred C57BL , Animals , Male , Cytokines/metabolism , Cytokines/genetics , Mice , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Disease Models, Animal , Humans , Fibrosis , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/drug therapy
10.
Microb Pathog ; 193: 106786, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971506

ABSTRACT

To better understand the interaction between attenuated vaccines and host antiviral responses, we used bioinformatics and public transcriptomics data to analyze the immune response mechanisms of host cells after canine distemper virus (CDV) infection in Vero cells and screened for potential key effector factors. In this study, CDV-QN-1 infect with Vero cells at an MOI of 0.5, and total RNA was extracted from the cells 24 h later and reverse transcribed into cDNA. Transcriptome high-throughput sequencing perform using Illumina. The results showed that 438 differentially expressed genes were screened, of which 409 were significantly up-regulated and 29 were significantly down-regulated. Eight differentially expressed genes were randomly selected for RT-qPCR validation, and the change trend was consistent with the transcriptomics data. GO and KEGG analysis of differentially expressed genes revealed that most of the differentially expressed genes in CDV-QN-1 infection in the early stage were related to immune response and antiviral activity. The enriched signaling pathways mainly included the interaction between cytokines and cytokine receptors, the NF-kappa B signaling pathway, the Toll-like receptor signaling pathway, and the NOD-like receptor signaling pathway. This study provides a foundation for further exploring the pathogenesis of CDV and the innate immune response of host cells in the early stage of infection.


Subject(s)
Distemper Virus, Canine , Gene Expression Profiling , Vaccines, Attenuated , Animals , Vero Cells , Chlorocebus aethiops , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Distemper Virus, Canine/genetics , Distemper Virus, Canine/immunology , Transcriptome , Signal Transduction , Computational Biology , High-Throughput Nucleotide Sequencing , Viral Vaccines/immunology , Viral Vaccines/genetics , Cytokines/metabolism , Cytokines/genetics , Distemper/virology , Distemper/genetics , Distemper/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , NF-kappa B/metabolism , NF-kappa B/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
11.
Signal Transduct Target Ther ; 9(1): 176, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39034318

ABSTRACT

Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-ß, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.


Subject(s)
Chemokines , Cytokines , Neoplasms , Signal Transduction , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/pathology , Signal Transduction/immunology , Signal Transduction/drug effects , Cytokines/immunology , Cytokines/genetics , Cytokines/metabolism , Chemokines/immunology , Chemokines/genetics , Chemokines/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/drug effects , Molecular Targeted Therapy
12.
Fish Shellfish Immunol ; 151: 109738, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971350

ABSTRACT

Antimicrobial peptides (AMPs) are an alternative to antibiotics for treatment and prevention of infections with a lower risk of bacterial resistance. Pituitary adenylate cyclase activating polypeptide (PACAP) is an outstanding AMP with versatile effects including antimicrobial activity and modulation of immune responses. The objective of this research was to study PACAP immunomodulatory effect on rainbow trout cell lines infected with Aeromonas salmonicida. PACAP from Clarias gariepinus (PACAP1) and a modified PACAP (PACAP5) were tested. RT-qPCR results showed that il1b and il8 expression in RTgutGC was significantly downregulated while tgfb expression was upregulated after PACAP treatment. Importantly, the concentration of IL-1ß and IFN-γ increased in the conditioned media of RTS11 cells incubated with PACAP1 and exposed to A. salmonicida. There was a poor correlation between gene expression and protein concentration, suggesting a stimulation of the translation of IL-1ß protein from previously accumulated transcripts or the cleavage of accumulated IL-1ß precursor. In-silico studies of PACAP-receptor interactions showed a turn of the peptide characteristic of PACAP-PAC1 interaction, correlated with the higher number of interactions observed with this specific receptor, which is also in agreement with the higher PACAP specificity described for PAC1 compared to VPAC1 and VPACA2. Finally, the in silico analysis revealed nine amino acids related to the PACAP receptor-associated functionality.


Subject(s)
Aeromonas salmonicida , Cytokines , Fish Proteins , Oncorhynchus mykiss , Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Aeromonas salmonicida/physiology , Oncorhynchus mykiss/immunology , Oncorhynchus mykiss/genetics , Cytokines/genetics , Cytokines/metabolism , Cell Line , Fish Diseases/immunology , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects , Catfishes/immunology , Catfishes/genetics , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Immunity, Innate/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics
13.
Medicine (Baltimore) ; 103(29): e38972, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39029049

ABSTRACT

Elevated levels of various cellular inflammatory markers have been observed in patients with endometriosis (EMs). However, a causal relationship between these markers and EMS has not been firmly established. This study aimed to assess the causality between cellular inflammatory markers and the onset of EMS using a bidirectional Mendelian randomization approach. Genetic associations for EMs were derived from the largest and most recent genome-wide association study (GWAS) involving 1937 EMS cases and 245,603 controls of European ancestry. Single nucleotide polymorphisms associated with 41 cellular cytokines and other systemic inflammatory regulators were identified from 8293 Finnish participants. Estimates were obtained using inverse-variance weighted, with sensitivity analyses conducted using MR-Egger, weighted median, and MR-PRESSO. Among the 41 systemic inflammatory regulators included in the analysis, none were associated with the risk of EMs. Elevated levels of IL-6 were associated with an increased risk of EMs (OR = 1.351, 95%CI = 1.015-1.797). Conversely, genetically predicted elevated levels of platelet-derived growth factor (PDGF-BB) were associated with a reduced risk of EMs (OR = 0.856, 95%CI = 0.742-0.987). Genetically predicted elevations in IL-6 may contribute to an increased risk of EMs, while elevated PDGF-BB levels appear protective, suggesting potential therapeutic targets for EMs. Other systemic inflammatory regulators seem unrelated to EMs risk, potentially representing downstream effects or consequences of shared factors between inflammation and EMs.


Subject(s)
Endometriosis , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Endometriosis/genetics , Endometriosis/blood , Female , Interleukin-6/genetics , Interleukin-6/blood , Inflammation/genetics , Inflammation/blood , Cytokines/blood , Cytokines/genetics , Finland/epidemiology , Genetic Predisposition to Disease
14.
Trop Anim Health Prod ; 56(6): 195, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963478

ABSTRACT

This experiment aimed to assess the regulatory effects of treatment with Balanites aegyptiaca fruit ethanol extract (BA-EE) on oxidant/antioxidant status, anti-inflammatory cytokines, and cell apoptosis gene expression in the abomasum of Haemonchus contortus-infected goats. Twenty goat kids were assigned randomly to four equal groups: (G1) infected-untreated, (G2) uninfected-BA-EE-treated, (G3) infected-albendazole-treated, (G4) infected-BA-EE-treated. Each goat in (G1), (G3), and (G4) was orally infected with 10,000 infective third-stage larvae. In the fifth week postinfection, single doses of albendazole (5 mg/kg.BW) and BA-EE (9 g/kg.BW) were given orally. In the ninth week postinfection, the animals were slaughtered to obtain abomasum specimens. The following oxidant/antioxidant markers were determined: malondialdehyde (MDA), glutathione (GSH), glutathione-S-transferase (GST), superoxide dismutase (SOD), catalase (CAT). The mRNA gene expression of cytokines (IL-3, IL-6, IL-10, TNF-α) and cell apoptosis markers (Bax, Bcl-2) were estimated. (G1) showed significantly reduced GSH content and GST and SOD activities but a markedly increased MDA level. (G3) and (G4) revealed a markedly lower MDA level with pronouncedly elevated GSH, SOD, and GST levels. The antioxidant properties of BA-EE were superior to those of albendazole. The mRNA gene expressions of IL-3, IL-6, IL-10, TNF-α, and Bax-2 were upregulated in (G1) but downregulated in (G3) and (G4). Bcl-2 and Bcl-2/Bax ratio expression followed a reverse course in the infected and both treated groups. We conclude that BA-EE treatment has a protective role in the abomasum of H. contortus-infected goats. This could be attributed to its antioxidant properties and ability to reduce pro-inflammatory cytokines and cell apoptosis.


Subject(s)
Abomasum , Antioxidants , Apoptosis , Cytokines , Goat Diseases , Goats , Haemonchiasis , Haemonchus , Plant Extracts , Animals , Goat Diseases/parasitology , Goat Diseases/drug therapy , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Cytokines/metabolism , Cytokines/genetics , Apoptosis/drug effects , Haemonchiasis/veterinary , Haemonchiasis/parasitology , Haemonchus/drug effects , Abomasum/parasitology , Antioxidants/metabolism , Anthelmintics/pharmacology , Anthelmintics/administration & dosage , Random Allocation , Ethanol , Gene Expression/drug effects , Albendazole/pharmacology , Albendazole/administration & dosage , Fruit/chemistry , Lamiaceae/chemistry , Male
15.
Zool Res ; 45(5): 961-971, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39016174

ABSTRACT

Bats, notable as the only flying mammals, serve as natural reservoir hosts for various highly pathogenic viruses in humans (e.g., SARS-CoV and Ebola virus). Furthermore, bats exhibit an unparalleled longevity among mammals relative to their size, particularly the Myotis bats, which can live up to 40 years. However, the mechanisms underlying these distinctive traits remain incompletely understood. In our prior research, we demonstrated that bats exhibit dampened STING-interferon activation, potentially conferring upon them the capacity to mitigate virus- or aging-induced inflammation. To substantiate this hypothesis, we established the first in vivo bat-mouse model for aging studies by integrating Myotis davidii bat STING ( MdSTING) into the mouse genome. We monitored the genotypes of these mice and performed a longitudinal comparative transcriptomic analysis on MdSTING and wild-type mice over a 3-year aging process. Blood transcriptomic analysis indicated a reduction in aging-related inflammation in female MdSTING mice, as evidenced by significantly lower levels of pro-inflammatory cytokines and chemokines, immunopathology, and neutrophil recruitment in aged female MdSTING mice compared to aged wild-type mice in vivo. These results indicated that MdSTING knock-in attenuates the aging-related inflammatory response and may also improve the healthspan in mice in a sex-dependent manner. Although the underlying mechanism awaits further study, this research has critical implications for bat longevity research, potentially contributing to our comprehension of healthy aging in humans.


Subject(s)
Aging , Chiroptera , Inflammation , Membrane Proteins , Animals , Female , Mice , Chiroptera/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism
16.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932231

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.


Subject(s)
Betacoronavirus 1 , Epithelial Cells , Gene Expression Profiling , Interferons , Animals , Swine , Epithelial Cells/virology , Epithelial Cells/immunology , Interferons/genetics , Interferons/metabolism , Interferons/immunology , Betacoronavirus 1/immunology , Betacoronavirus 1/genetics , Immunity, Innate , Virus Replication , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Cytokines/metabolism , Cytokines/genetics , Cytokines/immunology , Transcriptome , Respiratory Mucosa/virology , Respiratory Mucosa/immunology , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/genetics , Cells, Cultured , Deltacoronavirus
17.
Genes (Basel) ; 15(6)2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38927668

ABSTRACT

Tyrosine kinase inhibitor (TKI) drugs have significantly improved chronic myeloid leukemia (CML) outcomes. Neopeptides from CML cells may induce specific immune responses, which are crucial for deep molecular (DMR) and treatment-free remission (TFR). In this study of Ethiopian patients with CML (n = 162), the HLA alleles and single-nucleotide polymorphisms of five cytokines revealed significant associations with clinical outcomes. Clinically unfavorable outcomes correlated with HLA alleles A*03:01/02, A*23:17:01, B*57:01/02/03, and HLA-DRB4*01:01 (p-value = 0.0347, p-value = 0.0285, p-value = 0.037, and p-value = 0.0127, respectively), while HLA-DRB4*01:03:01 was associated with favorable outcomes (p-value = 0.0058). After assigning values for the 'low', 'intermediate', and 'high' gene expression of the SNPs' respective cytokine genes, Kaplan-Meier estimates for relapse-free survival, adjusted for age, treatment duration, and relapse risk among patients after the administration of TKIs, indicated that a gene expression ratio above the overall median of TNF-α, IL-6, and the combination of TGF-ß1/IL-10, IFNγ, and IL-6/IL-10 TGF-ß1 was correlated with a higher likelihood of treatment failure ((RR: 3.01; 95% CI: 1.1-8.3; p-value = 0.0261) and (RR: 2.4; 95% CI: 1.1-5.2; p-value = 0.022), respectively). Multi-SNPs, surpassing single-SNPs, and HLA allele polymorphisms showed promise in predicting outcomes of patients with CML during TKI treatment, prompting further exploration into their potential utility.


Subject(s)
Cytokines , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Alleles , Cytokines/genetics , HLA Antigens/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polymorphism, Single Nucleotide , Prognosis , /therapeutic use
18.
Biomed Khim ; 70(3): 156-160, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38940204

ABSTRACT

The cytokine profile of primary coronary artery endothelial cells cultivated in the presence of doxorubicin (2 µg/ml and 6 µg/ml) was evaluated using enzyme-linked immunosorbent assay and qPCR. Cultivation of cells in the presence of these concentrations of doxorubicin for 24 h, upregulated expression of the following genes: IL6 (by 2.30 and 2.66 times, respectively), IL1B (by 1.25 and 3.44 times), and CXCL8 (by 6.47 times and 6.42 times), MIF (2.34 and 2.28 times), CCL2 (4.22 and 3.98 times). Under these conditions the following genes were downregulated: IL10, IL1R2, TNF. Cultivation of cells in the presence of doxorubicin (2 µg/ml and 6 µg/ml) for 24 h also increased the secretion of IL-6.


Subject(s)
Coronary Vessels , Doxorubicin , Endothelial Cells , Interleukin-6 , Humans , Doxorubicin/pharmacology , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Interleukin-6/metabolism , Interleukin-6/genetics , Cells, Cultured , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Cytokines/metabolism , Cytokines/genetics , Gene Expression Regulation/drug effects , Interleukin-8/metabolism , Interleukin-8/genetics , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Interleukin-10/metabolism , Interleukin-10/genetics
19.
Front Immunol ; 15: 1339722, 2024.
Article in English | MEDLINE | ID: mdl-38903525

ABSTRACT

Background: A new aging biomarker epigenetic clock has been developed. There exists a close link between aging and gut microbiota, which may be mediated by inflammatory cytokines. However, the relationship between the epigenetic clock, gut microbiota, and the mediating substances is unclear. Methods: Two large genome-wide association meta-analyses were analyzed by two-sample Mendelian randomization. The results between gut microbiota and epigenetic clock were investigated using the four methods (Inverse variance weighted, MR-Egger, weighted median, MR-PRESSO). Genetic correlation was measured by Linked disequilibrium score regression (LDSC). The correctness of the study direction was checked by the Steiger test. Cochran's Q statistic and MR-Egger intercept were used as sensitivity analyses of the study. The two-step method was used to examine the mediating role of inflammatory cytokines. We use the Benjamini-Hochberg correction method to correct the P value. Results: After FDR correction, multiple bacterial genera were significantly or suggestively associated with four epigenetic clocks (GrimAge, HannumAge, IEAA, PhenoAge). And we detected several inflammatory factors acting as mediators of gut microbiota and epigenetic clocks. Conclusion: This study provides genetic evidence for a positive and negative link between gut microbiota and aging risk. We hope that by elucidating the genetic relationship and potential mechanisms between aging and gut microbiota, we will provide new avenues for continuing aging-related research and treatment.


Subject(s)
Cytokines , Epigenesis, Genetic , Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Gastrointestinal Microbiome/genetics , Humans , Cytokines/genetics , Cytokines/metabolism , Aging/genetics , Aging/immunology , Inflammation Mediators/metabolism , Polymorphism, Single Nucleotide
20.
Toxins (Basel) ; 16(6)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38922170

ABSTRACT

Snakebite envenoming is a neglected tropical disease that causes >100,000 deaths and >400,000 cases of morbidity annually. Despite the use of mouse models, severe local envenoming, defined by morbidity-causing local tissue necrosis, remains poorly understood, and human-tissue responses are ill-defined. Here, for the first time, an ex vivo, non-perfused human skin model was used to investigate temporal histopathological and immunological changes following subcutaneous injections of venoms from medically important African vipers (Echis ocellatus and Bitis arietans) and cobras (Naja nigricollis and N. haje). Histological analysis of venom-injected ex vivo human skin biopsies revealed morphological changes in the epidermis (ballooning degeneration, erosion, and ulceration) comparable to clinical signs of local envenoming. Immunostaining of these biopsies confirmed cell apoptosis consistent with the onset of necrosis. RNA sequencing, multiplex bead arrays, and ELISAs demonstrated that venom-injected human skin biopsies exhibited higher rates of transcription and expression of chemokines (CXCL5, MIP1-ALPHA, RANTES, MCP-1, and MIG), cytokines (IL-1ß, IL-1RA, G-CSF/CSF-3, and GM-CSF), and growth factors (VEGF-A, FGF, and HGF) in comparison to non-injected biopsies. To investigate the efficacy of antivenom, SAIMR Echis monovalent or SAIMR polyvalent antivenom was injected one hour following E. ocellatus or N. nigricollis venom treatment, respectively, and although antivenom did not prevent venom-induced dermal tissue damage, it did reduce all pro-inflammatory chemokines, cytokines, and growth factors to normal levels after 48 h. This ex vivo skin model could be useful for studies evaluating the progression of local envenoming and the efficacy of snakebite treatments.


Subject(s)
Cytokines , Necrosis , Skin , Humans , Skin/pathology , Skin/drug effects , Animals , Cytokines/metabolism , Cytokines/genetics , Snake Bites/pathology , Elapid Venoms/toxicity , Viper Venoms/toxicity , Inflammation/pathology , Inflammation/chemically induced , Viperidae , Chemokines/metabolism , Chemokines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...