Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Sci Rep ; 14(1): 15442, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965312

ABSTRACT

The human intestinal tract is colonized with microorganisms, which present a diverse array of immunological challenges. A number of antimicrobial mechanisms have evolved to cope with these challenges. A key defense mechanism is the expression of inducible antimicrobial peptides (AMPs), such as beta-defensins, which rapidly inactivate microorganisms. We currently have a limited knowledge of mechanisms regulating the inducible expression of AMP genes, especially factors from the host required in these regulatory mechanisms. To identify the host factors required for expression of the beta-defensin-2 gene (HBD2) in intestinal epithelial cells upon a bacterial challenge, we performed a RNAi screen using a siRNA library spanning the whole human genome. The screening was performed in duplicate to select the strongest 79 and 110 hit genes whose silencing promoted or inhibited HBD2 expression, respectively. A set of 57 hits selected among the two groups of genes was subjected to a counter-screening and a subset was subsequently validated for its impact onto HBD2 expression. Among the 57 confirmed hits, we brought out the TLR5-MYD88 signaling pathway, but above all new signaling proteins, epigenetic regulators and transcription factors so far unrevealed in the HBD2 regulatory circuits, like the GATA6 transcription factor involved in inflammatory bowel diseases. This study represents a significant step toward unveiling the key molecular requirements to promote AMP expression in human intestinal epithelial cells, and revealing new potential targets for the development of an innovative therapeutic strategy aiming at stimulating the host AMP expression, at the era of antimicrobial resistance.


Subject(s)
Epithelial Cells , Intestinal Mucosa , beta-Defensins , Humans , beta-Defensins/metabolism , beta-Defensins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Signal Transduction , Gene Expression Regulation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , RNA Interference
2.
J Nutr ; 154(7): 2244-2254, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38795745

ABSTRACT

BACKGROUND: Gut dysbiosis and increased intestinal permeability have been reported to precede type 1 diabetes-related autoimmunity. The role of gut inflammation in autoimmunity is not understood. OBJECTIVES: This study aimed to assess whether gut inflammation markers are associated with risk of islet autoimmunity and whether diet is associated with gut inflammation markers. METHODS: A nested case-control sample of 75 case children with islet autoimmunity and 88 control children was acquired from the Finnish Type 1 Diabetes Prediction and Prevention cohort. Diet was assessed with 3-d food records, and calprotectin and human ß-defensin-2 (HBD-2) were analyzed from stool samples at 6 and 12 mo of age. Conditional logistic regression analysis was used in a matched case-control setting to assess risk of autoimmunity. Analysis of variance, independent samples t test, and a general linear model were used in secondary analyses to test associations of background characteristics and dietary factors with inflammation markers. RESULTS: In unadjusted analyses, calprotectin was not associated with risk of islet autoimmunity, whereas HBD-2 in the middle (odds ratio [OR]: 3.23; 95% confidence interval [CI]: 1.03, 10.08) or highest tertile (OR: 3.02; 95% CI: 1.05, 8.69) in comparison to the lowest at 12 mo of age showed borderline association (P-trend = 0.063) with higher risk of islet autoimmunity. Excluding children with cow milk allergy in sensitivity analyses strengthened the association of HBD-2 with islet autoimmunity, whereas adjusting for dietary factors and maternal education weakened it. At age 12 mo, higher fat intake was associated with higher HBD-2 (ß: 0.219; 95% CI: 0.110, 0.328) and higher intake of dietary fiber (ß: -0.294; 95% CI: -0.510, -0.078), magnesium (ß: -0.036; 95% CI: -0.059, -0.014), and potassium (ß: -0.003; 95% CI: -0.005, -0.001) with lower HBD-2. CONCLUSIONS: Higher HBD-2 in infancy may be associated with higher risk of islet autoimmunity. Dietary factors play a role in gut inflammatory status.


Subject(s)
Autoimmunity , Biomarkers , Diabetes Mellitus, Type 1 , Diet , Islets of Langerhans , Leukocyte L1 Antigen Complex , beta-Defensins , Humans , Case-Control Studies , Finland , Female , Male , Leukocyte L1 Antigen Complex/analysis , Diabetes Mellitus, Type 1/immunology , Infant , Islets of Langerhans/immunology , Risk Factors , Inflammation , Feces/chemistry
3.
BMC Pediatr ; 24(1): 183, 2024 Mar 16.
Article in English | MEDLINE | ID: mdl-38491401

ABSTRACT

BACKGROUND: This study aimed to assess the diagnostic potential of serum intestinal fatty acid-binding protein (I-FABP), fecal calprotectin (FC), and fecal human ß-defensin 2 (hBD2) in predicting necrotizing enterocolitis (NEC) in preterm infants. METHODS: A prospective cohort of neonates with a gestational age < 32 weeks, suspected of NEC, was enrolled between June 2021 and December 2022. Serum I-FABP, FC, and fecal hBD2 levels were measured upon NEC suspicion, and diagnosis was confirmed through radiological examination or surgical intervention. Diagnostic precision of serum I-FABP, FC, and fecal hBD2 was assessed using a logistic regression model with multiple variables. RESULTS: The study included 70 neonates (45 males, 25 females), with 30 developing NEC (40% Stage III, n = 12; 60% Stage II, n = 18) and 40 in the control group. NEC patients exhibited significantly higher serum I-FABP and FC levels (4.76 ng/mL and 521.56 µg/g feces, respectively) than those with other diagnoses (1.38 ng/mL and 213.34 µg/g feces, respectively; p ˂ 0.05 for both biomarkers). Stage II NEC neonates showed elevated fecal hBD2 levels (376.44 ng/g feces) than Stage III NEC neonates and controls (336.87 ng/g and 339.86 ng/g feces, respectively; p ˂ 0.05). No such increase was observed in infants progressing to Stage III NEC. Using a serum I-FABP threshold of > 2.54 ng/mL yielded 76.7% sensitivity, 87.5% specificity, 82.1% positive predictive value (PPV), and 83.3% negative predictive value (NPV). For FC (cutoff > 428.99 µg/g feces), corresponding values were 76.7% sensitivity, 67.5% specificity, 63.9% PPV, and 79.4% NPV. CONCLUSION: Serum I-FABP and FC levels are valuable for early NEC detection and provide insights into disease severity. Low fecal hBD2 levels suggest an inadequate response to luminal bacteria, potentially rendering these infants more susceptible to NEC development or exacerbation.


Subject(s)
Enterocolitis, Necrotizing , Infant, Newborn, Diseases , beta-Defensins , Male , Infant , Female , Infant, Newborn , Humans , Infant, Premature , Enterocolitis, Necrotizing/metabolism , Leukocyte L1 Antigen Complex/metabolism , beta-Defensins/metabolism , Prospective Studies , Fatty Acid-Binding Proteins , Feces , Biomarkers/metabolism
4.
Article in English | MEDLINE | ID: mdl-38376819

ABSTRACT

Human intestinal epithelial cells (IECs) play an important role in maintaining gut homeostasis by producing antimicrobial peptides (AMPs). Bacillus subtilis, a commensal bacterium, is considered a probiotic. Although its protective effects on intestinal health are widely reported, the key component of B. subtilis responsible for its beneficial effects remains elusive. In this study, we tried to identify the key molecules responsible for B. subtilis-induced AMPs and their molecular mechanisms in a human IEC line, Caco-2. B. subtilis increased human beta defensin (HBD)-2 mRNA expression in a dose- and time-dependent manner. Among the B. subtilis microbe-associated molecular patterns, lipoprotein (LPP) substantially increased the mRNA expression and protein production of HBD-2, whereas lipoteichoic acid and peptidoglycan did not show such effects. Those results were confirmed in primary human IECs. In addition, both LPP recognition and HBD-2 secretion mainly took place on the apical side of fully differentiated and polarized Caco-2 cells through Toll-like receptor 2-mediated JNK/p38 MAP kinase/AP-1 and NF-κB pathways. HBD-2 efficiently inhibited the growth of the intestinal pathogens Staphylococcus aureus and Bacillus cereus. Furthermore, LPPs pre-incubated with lipase or proteinase K decreased LPP-induced HBD-2 expression, suggesting that the lipid and protein moieties of LPP are crucial for HBD-2 expression. Q Exactive Plus mass spectrometry identified 35 B. subtilis LPP candidates within the LPP preparation, and most of them were ABC transporters. Taken together, these results suggest that B. subtilis promotes HBD-2 secretion in human IECs mainly with its LPPs, which might enhance the protection from intestinal pathogens.

5.
Int J Health Sci (Qassim) ; 18(1): 17-23, 2024.
Article in English | MEDLINE | ID: mdl-38188898

ABSTRACT

Objective: The objective of this study was to assess the periodontal health status of individuals with lung cancer in the North Indian population. In addition, the study aimed to determine the levels of human beta-defensin2 (Hbd-2) in the gingival crevicular fluid (GCF) and serum samples collected from the participants. Methods: The study consisted of a total of 90 participants, who were categorized into three groups: Group 1 included 30 healthy individuals, Group 2 comprised 30 patients with chronic periodontitis, and Group 3 involved 30 patients diagnosed with both lung cancer and chronic periodontitis. Various periodontal parameters, including plaque index, gingival index, probing pocket depth, and clinical attachment level (CAL), were assessed in addition to the analysis of human beta defensin2 levels in both the GCF and serum samples of all participants. Results: The study results revealed that all clinical parameters assessed were higher in Group 3 compared to both Group 2 and Group 1. Specifically, the levels of hBD-2 in the GCF were measured as 52.29 ± 46.41 pg/mL in Group 1, 27.15 ± 28.76 pg/mL in Group 2, and 86.01 ± 68.82 pg/mL in Group 3. When comparing the hBD-2 levels in serum, the values were found to be 813.72 ± 269.43 pg/mL in Group 1, 591.50 ± 263.91 pg/mL in Group 2, and 1093.04 ± 674.55 pg/mL in Group 3. These intergroup comparisons indicate variations in hBD-2 levels among the different groups. Conclusions: The study findings demonstrated significantly higher clinical and biochemical markers in patients with both lung cancer and chronic periodontitis, in comparison to individuals with chronic periodontitis alone and healthy participants. These results suggest that Hbd-2 could potentially serve as a valuable diagnostic biomarker for identifying and distinguishing individuals with both lung cancer and chronic periodontitis.

6.
Microb Pathog ; 186: 106469, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38070628

ABSTRACT

ß-defensin 2 (BD2) is a small cationic peptide that exerts a critical role in host defense against bacterial infections. Here, we aimed to investigate the role of BD2 in protecting against acute urinary tract infection (AUTI) caused by Escherichia coli (UPEC). Here, LPS-induced human urinary bladder epithelial cell (HCV-29) model and UPEC-induced mice model were used for assessing AUTI. Visceral organ lesions of mice following treatment was assessed by HE staining. Cell viability was determined by CCK-8 assay. Permeability in HCV-29 cells was analyzed in Transwell assay. Expression of inflammatory factors (IL-1ß, IL-6, and TNF-α) was measured by ELISA assay. The levels of BD2, ß-catenin and tight-junction proteins (ZO-1, Occludin, and Claudin-1) were detected by RT-qPCR, western blotting, immunofluorescence or immunohistochemistry. Our results showed that BD2 was lowly expressed and ß-catenin showed the reverse trend in response to bacterial infection in vitro and in vivo. BD2 overexpression alleviated the decreased cell viability, increased cell permeability, upregulation of inflammatory factors, downregulation of tight-junction protein and high ß-catenin expression in LPS-induced HCV-29 cells, which may contribute to the negative regulation of ß-catenin expression. Furthermore, BD2 overexpression attenuated the bacterial infection of tissues, high levels of inflammatory factors and ß-catenin, and low levels of tight-junction proteins in mice stimulated with UPEC. This study showed that BD2 played a crucial role in protecting against AUTI caused by gram-negative bacteria through suppressing ß-catenin expression. Targeting BD2 may provide a potential therapeutic approach for the prevention and treatment of AUTI.


Subject(s)
Escherichia coli Infections , Hepatitis C , Urinary Tract Infections , beta-Defensins , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , beta-Defensins/genetics , beta-Defensins/metabolism , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Lipopolysaccharides/toxicity , Tight Junction Proteins/metabolism
7.
Sci Total Environ ; 905: 167201, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-37734607

ABSTRACT

Aflatoxin B1 (AFB1) is a highly toxic fungal toxin that causes severe damage to animal intestines. Porcine beta-defensin-2 (pBD-2) is a well-studied antimicrobial peptide in pigs that can protect animal intestines and improve productivity. This study aimed to investigate the molecular mechanisms of pBD-2 in alleviating AFB1-induced oxidative stress and intestinal mucosal damage using porcine intestinal epithelial cells (IPEC-J2 cells) and Kunming (KM) mice. The maximum destructive concentration of AFB1 for IPEC-J2 cells and the optimal therapeutic concentration of pBD-2 were determined by CCK-8 and RT-qPCR. We then investigated the oxidative stress and intestinal damage induced by AFB1 and the alleviating effect of pBD-2 by detecting changes of reactive oxygen species (ROS), inflammatory cytokines, tight junction proteins (TJPs) and mucin. Finally, the molecular mechanism of pBD-2 mitigates AFB1-induced oxidative stress and intestinal mucosal damage were explored by adding ROS and Erk1/2 pathway inhibitors to comparative analysis. In vivo, the therapeutic effect of pBD-2 on AFB1-induced intestinal damage was analyzed from aspects such as average daily gain (ADG), pathological damage, inflammation, and mucosal barrier in KM mice. The study found that low doses of pBD-2 promoted cell proliferation and prevented AFB1-induced cell death, and pBD-2 significantly restored the feed conversion rate and ADG of KM mice reduced by long-term exposed AFB1. Increasing the intracellular ROS and the expression and phosphorylation of Erk1/2, AFB1 promoted inflammation by altering inflammatory cytokines TNF-α, IL-1ß, IL-6, and IL-8, and disrupted the mucosal barrier by interfering with Claudin-3, Occludin, and MUC2, while pBD-2 significantly reduced ROS and decreased the expression and phosphorylation of Erk1/2 to restored their expression to alleviate AFB1-induced oxidative stress and intestinal mucosal damage in IPEC-J2 cells and the small intestine of mice.


Subject(s)
Animals, Outbred Strains , beta-Defensins , Mice , Swine , Animals , Reactive Oxygen Species/metabolism , Aflatoxin B1/toxicity , Cell Line , Signal Transduction , Cytokines , Inflammation
8.
Protein J ; 42(4): 399-407, 2023 08.
Article in English | MEDLINE | ID: mdl-37291459

ABSTRACT

COVID-19 is a disease that have affected the entire world, and it continues to spread with new variants. A patient's innate immune system plays a critical role in the mild and severe transition of COVID-19. Antimicrobial peptides (AMPs), which are important components of the innate immune system, are potential molecules to fight pathogenic bacteria, fungi, and viruses. Human ß-defensin 2 (hBD-2), a 41-amino-acid antimicrobial peptide, is one of the defensins inducibly expressed in the skin, lungs, and trachea in humans. In this study, it was aimed to investigate the interaction of hBD-2 produced recombinantly in Pichia pastoris with the human angiotensin-converting enzyme 2 (ACE-2) under in vitro conditions. First, hBD-2 was cloned in P. pastoris X-33 via the pPICZαA vector, a yeast expression platform, and its expression was confirmed by SDS-PAGE, western blotting, and qRT-PCR. Then, the interaction between recombinant hBD-2 and ACE-2 proteins was revealed by a pull-down assay. In light of these preliminary experiments, we suggest that the recombinantly produced hBD-2 may be protective against SARS-CoV-2 and be used as a supplement in treatment. However, current findings need to be supported by cell culture studies, toxicity analyses, and in vivo experiments.


Subject(s)
COVID-19 , beta-Defensins , Humans , beta-Defensins/genetics , beta-Defensins/pharmacology , beta-Defensins/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Pichia/genetics , Pichia/metabolism
9.
Biomed Pharmacother ; 161: 114540, 2023 May.
Article in English | MEDLINE | ID: mdl-36934557

ABSTRACT

Diabetic wound (DW) is the most devastating complication resulting in significant mortality and morbidity in diabetic patients. The standard treatment of DW care fails to address the prerequisites of treating DW owing to its multifactorial pathophysiology. Henceforth, developing a single treatment strategy to handle all the loopholes may effectively manage DW. The objective of the current study was to formulate Human beta defensin-2 (HBD-2) loaded Poly (lactic-co-glycolic acid) (PLGA) nanoparticle impregnated in collagen/chitosan (COL-CS) composite scaffolds for the accelerated healing of DW. Upon investigation, the developed biodegradable crosslinked scaffold possesses low matrix degradation, optimum porosity, and sustained drug release than the non-crosslinked scaffold. In vitro studies revealed that the HBD-2 COL-CS scaffold was biocompatible and accelerated cell migration and angiogenesis. The HBD-2 COL-CS scaffold showed significant antimicrobial activity in S. aureus, E. coli, and P. aeruginosa. The in vivo studies revealed that the HBD-2 COL-CS treated group accelerated healing compared to those in COL-CS and control groups. The ELISA results indicated a significant decrease in MMP-9, TNF-α, MPO, NAG, and NO with an increase in IL-10 in HBD-2 COL-CS treated group. The accelerated healing in HBD-2 COL-CS treated group might be due to the synergistic effects of PLGA (collagen synthesis and deposition and positive angiogenic effect), HBD-2 (anti-inflammatory, antibacterial, positive angiogenic effect, cell proliferation, and migration), COL (established wound healer and stabilizer) and CS (antibacterial, controlled drug release).


Subject(s)
Chitosan , Diabetes Mellitus , Nanoparticles , beta-Defensins , Humans , Tissue Scaffolds , Staphylococcus aureus , Escherichia coli , Collagen/pharmacology , Anti-Bacterial Agents/pharmacology
10.
J Clin Med ; 12(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902728

ABSTRACT

(1) Background: Atopic dermatitis is one of the most common inflammatory skin diseases characterized by T helper (Th) 2 and Th22 cells producing interleukin (IL)-4/IL-13 and IL-22, respectively. The specific contribution of each cytokine to the impairment of the physical and the immune barrier via Toll-like receptors (TLRs) is poorly addressed concerning the epidermal compartment of the skin. (2) Methods: The effect of IL-4, IL-13, IL-22, and the master cytokine IL-23 is evaluated in a 3D model of normal human skin biopsies (n = 7) at the air-liquid interface for 24 and 48 h. We investigated by immunofluorescence the expressions of (i) claudin-1, zonula occludens (ZO)-1 filaggrin, involucrin for the physical barrier and (ii) TLR2, 4, 7, 9, human beta-defensin 2 (hBD-2) for the immune barrier. (3) Results: Th2 cytokines induce spongiosis and fail in impairing tight junction composition, while IL-22 reduces and IL-23 induces claudin-1 expression. IL-4 and IL-13 affect the TLR-mediated barrier largely than IL-22 and IL-23. IL-4 early inhibits hBD-2 expression, while IL-22 and IL-23 induce its distribution. (4) Conclusions: This experimental approach looks to the pathogenesis of AD through molecular epidermal proteins rather than cytokines only and paves the way for tailored patient therapy.

11.
Odontology ; 111(4): 830-838, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36745267

ABSTRACT

ß-defensin 2 (BD-2), an antimicrobial peptide (AMP), is expressed by oral epithelial cells and plays an important role in innate immunity of the oral cavity. Cell-free protein synthesis (CFPS) systems have been studied for the synthesis of various proteins, however, the synthesis of BD-2 by a CFPS system has not been extensively explored. Liposomes have been developed as tools for drug delivery. A delivery of liposome-encapsulated AMP to oral epithelium may be useful to prevent oral infectious diseases. In the present study, we investigated the antimicrobial activity of the BD-2 protein, artificially synthesized using a CFPS system and encapsulated in liposomes. BD-2 protein was artificially synthesized using template DNA and a reconstituted CFPS system and was identified by western blotting. Bilayer liposomes were prepared using 1,2-dioleoyl-sn-glycero-3-phospho-choline and 3-sn-phosphatidylcholine from egg yolk. The artificially synthesized BD-2 was encapsulated in liposomes, collected by ultrafiltration, and detected by western blotting. Human oral epithelial cells were cultured with the liposome-encapsulated BD-2 and the concentration of BD-2 in the cell lysate of the culture with the synthesized BD-2 was higher than that of the control cultures. The antimicrobial activity of the synthesized BD-2 was investigated by an adhesion assay of Porphyromonas gingivalis to oral epithelial cells. The artificially synthesized BD-2 and its liposome significantly inhibited adhesion of P. gingivalis to oral epithelial cells. These results suggest that artificially synthesized BD-2 and liposome-encapsulated BD-2 show antimicrobial activity and can potentially play a role in oral healthcare for periodontal diseases.


Subject(s)
Anti-Infective Agents , beta-Defensins , Humans , Porphyromonas gingivalis , Liposomes/pharmacology , Liposomes/metabolism , beta-Defensins/pharmacology , beta-Defensins/metabolism , Epithelial Cells/metabolism , Proteins/metabolism , Anti-Infective Agents/metabolism
12.
Lett Appl Microbiol ; 76(1)2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36688785

ABSTRACT

The efficacy of Lactobacillus as an antibiotic substitute has been investigated as one of the potential strategies to prevent Salmonella infection in poultry. The purpose of this study was to explore the antibacterial activity of Lactobacillus fermentum 1.2133 (Lact. fermentum 1.2133) against Salmonella pullorum CVCC533 (Salm. pullorum CVCC533) and its effect on chickens infected with Salm. pullorum CVCC533. Results showed that Lact. fermentans 1.2133 has antibacterial activity against Salm. pullorum CVCC533 and the cell-free fermentation supernatant of Lact. fermentum 1.2133 had a bactericidal effect on the bacteria in the Salm. pullorum CVCC533 biofilm by significantly reducing the number of Salmonella and aerobic bacteria in the chicken duodenum, ileum, and cecum, including Escherichia shigella (P < 0.05), improved the species abundance of Lactobacilli (P < 0.05). The damage to the chicken intestine by Salm. pullorum CVCC533 was reduced as the expression of avian beta-defensin 2 (AvBD2) mRNA in chicken small intestine was increased (P < 0.05). The results showed that Lact. fermentum 1.2133 had the potential to be a probiotic for poultry due to its regulation of intestinal AvBD2 mRNA as well as its intestinal flora.


Subject(s)
Limosilactobacillus fermentum , Poultry Diseases , Probiotics , Salmonella Infections, Animal , Animals , Chickens/microbiology , Lactobacillus/physiology , Salmonella , Poultry , Anti-Bacterial Agents
13.
Front Pediatr ; 10: 1062798, 2022.
Article in English | MEDLINE | ID: mdl-36582510

ABSTRACT

Background and purpose: Necrotizing enterocolitis (NEC) is a critical gastrointestinal disease. We aim to explore the value of fecal human ß-defensin 2 (HBD-2), Claudin-3, high-mobility group box-1 protein (HMGB-1), and resistin-like molecule ß (Relmß) as well as some laboratory metrics to predict the deterioration of NEC. Methods: Infants diagnosed with NEC at Stage II were enrolled in our study. Those who progressed to Stage III were included in the Stage III group and the rest were included in the Stage II group. Clinical data and laboratory metrics of the infants were collected. Fecal samples of HBD2, HMGB-1, Claudin-3, and Relmß collected during their enrollment were determined by using enzyme-linked immunosorbent assay (ELISA) kits. Student's t-test, the Mann-Whitney U test, the chi-square test, receiver operating characteristic (ROC), and logistic regression analysis were performed. Results: Sixty infants diagnosed with NEC at Stage II were enrolled in our study, with 27 in the Stage III group (n = 27) and 33 in the Stage II group (n = 33). Although many of these NEC cases were late preterm and term infants, the infants in the Stage III group had a lower gestational age (P < 0.05). The incidence of gestational diabetes mellitus, peritonitis, intestinal adhesion, and sepsis was higher and more infants in the Stage III group underwent surgeries (P < 0.05). The levels of HBD-2 and Claudin-3 were higher and neutrophil count was lower in the Stage III group than in the Stage II Group, and the area under the curve (AUC) was 0.754, 0,755, and 0.666, respectively (P < 0.05). HBD-2 ≥ 1649.02 ng/g and Claudin-3 ≥ 2488.71 pg/g were included in the multivariate stepwise logistic regression analysis (P < 0.05), and the AUC of the model was 0.805 (95% CI: 0.688-0.922). Conclusion: Fecal HBD-2 and Claudin-3 may be potential biomarkers to predict the deterioration of NEC from Stage II to Stage III.

14.
BMC Immunol ; 23(1): 53, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36324077

ABSTRACT

BACKGROUND: Macrophages are mononuclear CD34+ antigen-presenting cells of defense mechanism and play dual roles in tumor burden. The immunomodulatory and their antitumor function of ß-defensin 2 is still unclear, despite the accumulating evidence of the response in infection. So, the aim of present study is to elucidate the role of ß-defensin 2 on the level of ROS, cytokines, chemokine expression in macrophages and antitumor function in breast cancer. METHOD: Swiss albino mice were used to harvest PEC macrophages and C127i breast cancer cells line for tumor model was used in this study. Macrophages were harvested and characterized by flow-cytometry using F4/80 and CD11c antibodies. MTT was performed to estimate cytotoxicity and dose optimization of ß-defensin 2. Oxidative stress was analyzed by H2O2 and NO estimation followed by iNOS quantified by q-PCR. Cytokines and chemokines estimation was done using q-PCR. Co-culture experiment was performed to study anti-tumor function using PI for cell cycle, Annexin -V and CFSE analysis for cell proliferation. RESULTS: PEC harvested macrophages were characterized by flow-cytometry using F4/80 and CD11c antibodies with the purity of 8% pure population of macrophages. It was found that 99% of cells viable at the maximum dose of 100 ng/ml of ß-defensin 2 in MTT. Levels of NO and H2O2 were found to be decreased in ß-defensin 2 as compared to control. Expression of cytokines of IFN-γ, IL-1α, TNF-α, TGF-ßwas found to be increased while IL-3 was decreased in ß-defensin 2 group as compared to control. Levels of chemokines CXCL-1, CXCL-5 and CCL5 increased in treated macrophages while CCL24 and CXCL-15 expression decreased. Adhesion receptor (CD32) and fusion receptor (CD204) were decreased in the ß-defensin 2 group as compared to control. Anti-tumor experiment was performed using co-culture experiment apoptosis (Annexin-V) was induced, cell cycle arrest in phage and cell proliferation of C127i cells was decreased. CONCLUSION: This is the first report of ß-defensin 2 modulates macrophage immunomodulatory and their antitumor function in breast cancer. ß-defensin 2 as a new therapeutic target for immunotherapy as an adjuvant in vaccines.


Subject(s)
Neoplasms , beta-Defensins , Animals , Mice , beta-Defensins/metabolism , beta-Defensins/pharmacology , Hydrogen Peroxide , Macrophages , Cytokines/metabolism , Chemokines/metabolism , Chemokines/pharmacology , Annexins/metabolism , Annexins/pharmacology , Neoplasms/metabolism
15.
Trop Life Sci Res ; 33(2): 75-90, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35966270

ABSTRACT

Probiotic Lactobacillus reuteri has positive effects on health through inhibiting pathogenic bacteria and the ability to reduce inflammation. This study investigates the ability of reuterin isolated from L. reuteri Indonesian strain for increasing mRNA expression of interleukin (IL)-8 and human beta-defensin (hBD)-2 gene by epithelial cells, after exposure to oral bacteria. L. reuteri isolated from Indonesian's saliva, and species was confirmed by PCR, using 16S rRNA specific gene. To produce reuterin, the isolate was mixed in glycerol-containing MRS broth. Reuterin molecule's weight was counted by SDS-PAGE. Streptococcus mutans ATCC-25175 and Porphyromonas gingivalis ATCC-33277 were put in water (80°C) for 30 min, and each killed bacterial (107 CFU/mL) was inoculated into HaCat cell line (105 cell/mL). Reuterin was added in different concentrations (100%, 50%, 25%, 12,5%) and different incubation time at 37°C, 5% CO2. RNA was extracted, and a reverse transcription procedure was performed to obtain cDNA. Subsequently, a quantitative PCR method was performed to analyse the transcription level of IL-8 and HBD-2 mRNA expressed by inflamed HaCat cells. All results were statistically analysed by ANOVA test. PCR assays showed that clinical isolates were L. reuteri. Quantitative PCR results showed reuterin decreased the expression of IL-8 and increased the expression of hBD-2 in all concentrations and time periods set in this study (p < 0.05). Reuterin isolated from L. reuteri Indonesian strain increased expression of human beta defensin-2 as antimicrobial peptide and may be useful in combating inflammation.

16.
Front Pediatr ; 10: 873793, 2022.
Article in English | MEDLINE | ID: mdl-35733815

ABSTRACT

Background: Celiac disease (CD) is an immune-mediated disorder of the gut in which innate and adaptive responses are involved. Antimicrobial peptides (AMPs) constitute an arsenal of innate immunity regulators of paramount importance in the gut. However, the role of AMPs in CD is unclear. Aims: To evaluate the levels of fecal ß-defensin-2, fecal calprotectin (FC), and antibodies against bactericidal/permeability-increasing protein (BPI) in the serum of children with active CD and to compare them with those of healthy controls (HCs). Methods: We examined 76 children with recently diagnosed CD between the age of 2-10 years (average age: 6.1 ± 1.2 years) and 32 HC (average age: 6.2 ± 3.8 years) in this study. We evaluated the level of fecal ß-defensin-2 and FC levels in coprofiltrates, and the level of anti-BPI antibodies in blood serum. Correlation relationships between the parameters were assessed according to Pearson correlation coefficient. Results: Fecal ß-defensin-2 concentration was greater in the CD group than in HC group, amounting to 99.6 ± 15.5 ng/mL and 64.0 ± 2.4 ng/mL, respectively (p < 0.02). The level of FC in the CD children was 35.4 ± 8.1 µg/g, while that in the control group was 19.1 ± 1.1 µg/g, (p < 0.05), representing a slightly increase. The concentration of anti-BPI antibodies in the CD and HC groups was 35.9 ± 10.1 U/mL and 5.2 ± 3.2 U/mL, respectively (p < 0.002). There was a strong and direct correlation between fecal ß-defensin-2 and FC (r = 0.69), as well as a direct but weak relationship between fecal ß-defensin-2 and anti-BPI antibodies (r = 0.35). Conclusions: Our data reinforce that fecal ß-defensin-2 and anti-BPI antibodies are greatly increased in patients with active CD. These biomarkers may be components of epithelial innate immunity in the intestine, with each having a distinct functional role in intestinal6 mucosal defense.

17.
Am J Transl Res ; 14(5): 3286-3291, 2022.
Article in English | MEDLINE | ID: mdl-35702098

ABSTRACT

OBJECTIVE: To examine the correlation between salivary human ß defensin-2 (HBD-2) and LL-37 expression levels and blood glucose in relationship to periodontal status in patients with type 2 diabetes mellitus (T2DM). METHODS: The trial is available at: https://clinicaltrials.gov/ with ClinicalTrials.gov Identifier: NCT03512675. A total of 89 patients with T2DM with chronic periodontitis (CP) enrolled in our hospital from January 2020 to December 2020 were selected. According to the degree of glycemic control and CP, the patients were randomly divided into four groups, namely the good glycemic control and mild CP group (n=26), good glycemic control with moderate to severe CP group (n=24), poor glycemic control with mild CP group (n=21), and poor glycemic control with moderate to severe CP group (n=18). The periodontal clinical parameters, blood glucose indicators, and saliva HBD-2 and LL-37 expression levels were determined. RESULTS: The expression levels of HBD-2 and LL-37 in the saliva of T2DM patients with moderate to severe CP and poor blood sugar control were significantly increased (P<0.05). Saliva HBD-2 and LL-37 levels were positively correlated with probing depth, clinical attachment loss, plaque index, and glycosylated hemoglobin. There was a synergistic interaction between blood glucose, periodontal status, and saliva HBD-2, LL-37 levels (P<0.05). CONCLUSION: There is a positively correlated relationship between blood glucose and periodontal status with salivary HBD-2 and LL-37 levels.

18.
World Allergy Organ J ; 15(5): 100633, 2022 May.
Article in English | MEDLINE | ID: mdl-35600835

ABSTRACT

Background: This study aimed to investigate whether fecal human beta-defensins (HBD)-2 and eosinophil cationic protein (ECP) expression in preterm infants are associated with allergic disease development by age 2 years. Methods: Preterm infants' stool samples were collected at the age of 6 and 12 months postnatally. Information regarding medication exposure histories (antibiotics, antipyretics, probiotics) and physician-diagnosed allergic diseases was obtained using age-specific questionnaires and medical records. We compared the 6-month and 12-month fecal HBD-2 and ECP concentrations between the medication exposure and non-exposure group, respectively, and between children who developed allergic diseases and those who did not by 2 years of age. Univariate and multivariable logistic regression analyses were performed to investigate independent variables related to physician-diagnosed allergic diseases by 2 years of age. Results: Seventy-four preterm infants (gestational age, 31-36 weeks) were included. Fecal HBD-2 levels were significantly increased at 12 months of age among children who developed allergic diseases compared to those who did not (37.18 ± 11.80 ng/g vs. 8.56 ± 4.33 ng/g, P = 0.011). This association was more apparent among allergic children given antibiotics (50.23 ± 16.15 ng/g vs. 9.75 ± 7.16 ng/g, P = 0.008) or antipyretics (46.12 ± 14.22 ng/g vs. 10.82 ± 6.81 ng/g, P = 0.018) during the first year, whereas among allergic children who were previously not exposed to antibiotics or antipyretics, the differences were not significant. Results of the multivariable logistic regression analysis indicated that HBD-2 concentration in 12-month stools was an independent indicator associated with physician-diagnosed allergic diseases by 2 years of age (adjusted odds ratio: 1.03 [95% confidence interval: 1.00-1.05], P = 0.036). Our data revealed a lack of association between fecal ECP and allergic diseases. Conclusions: We found that preterm infants who expressed high fecal HBD-2 at 12 months of age were associated with physician-diagnosed allergic diseases by the age of 2 years. Further studies are needed to determine the role of fecal HBD-2 in the development of allergic diseases.

19.
Int J Biol Macromol ; 208: 890-900, 2022 May 31.
Article in English | MEDLINE | ID: mdl-35364205

ABSTRACT

pBD2 is one of the porcine beta defensins with broad antimicrobial activity, and plays an important role in immune regulation. However, the activities and mechanisms of pBD2 regulating host resistance to Escherichia coli infection are unclear. In this study, the immunomodulatory activity and mechanisms of recombinant pBD2 against Escherichia coli infection were explored in IPEC-J2 cells. Recombinant pBD2 had no obvious effect on the growth of cells below 80 µg/mL, however, it reduced the number of E. coli adhering to cells. Furthermore, pBD2 restored the abnormal expression of ZO-1 and occludin in cells challenged with E. coli. pBD2 treatment also reduced cell apoptosis and decreased the expression of the apoptosis-related genes Cox-2 and Caspase-3, and decreased the expression of the pro-inflammatory IL-6, IL-8, IL-1α and TNF-α, and Cxcl2 and Ccl20. pBD2 also reduced the expression of TAK1, and inhibited the phosphorylation of NF-κB p65 following E. coli infection. In addition, pBD2 was localized in the cytoplasm. Collectively, pBD2 appeared to penetrate cells and alleviate inflammatory responses via the TAK1-NF-κB signaling pathway. Our results revealed the immunomodulatory activity of recombinant pBD2 against E. coli and provided insights into the molecular mechanisms that protected cells from E. coli infection.


Subject(s)
Escherichia coli Infections , beta-Defensins , Animals , Epithelial Cells , Escherichia coli , Escherichia coli Infections/metabolism , Intestinal Mucosa/metabolism , NF-kappa B/metabolism , Signal Transduction , Swine , beta-Defensins/genetics , beta-Defensins/pharmacology
20.
Antiviral Res ; 201: 105292, 2022 05.
Article in English | MEDLINE | ID: mdl-35341807

ABSTRACT

Swine influenza virus (SIV) not only brings about great economic losses on the global pig industry, it also poses a significant threat to the public health for its interspecies transmission capacity. Porcine ß-defensin 2 (PBD-2) is a host defense peptide and our previous study has shown that PBD-2 inhibits proliferation of enveloped pseudorabies virus both in vitro and in transgenic (TG) mice. The aim of this study is to investigate the possible anti-SIV ability of PBD-2 in a TG pig model created in our previous study. The in-contact challenge trial demonstrated that overexpression of PBD-2 in pigs could efficiently alleviate SIV-associated clinical signs. The SIV titers quantified by EID50 in lung tissues of infected TG pigs were significantly lower than that of wild-type littermates. In vitro, the cell viability assay revealed that PBD-2 mainly interfered with viral entry and post-infection stages. It was further confirmed that PBD-2 could enter porcine tracheal epithelial cells. The proteins interacting with PBD-2 inside host cells were identified with immunoprecipitation and the pathways involved were analyzed. Results showed that PBD-2 could interact with pro-apoptotic solute carrier family 25 member 4 (SLC25A4), also known as adenine nucleotide translocase 1, and thereby inhibited SIV-induced cell apoptosis. The molecular docking analysis suggested that PBD-2 interacted with porcine SLC25A4 mainly through strong hydrogen binding, with the predicted binding affinity being -13.23 kcal/mol. Altogether, these indicate that PBD-2 protects pigs against SIV infection, which may result from its role as a SLC25A4 blocker to alleviate cell apoptosis, providing a novel therapeutic and prophylactic strategy of using PBD-2 to combat SIV.


Subject(s)
Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Swine Diseases , beta-Defensins , Animals , Apoptosis , Humans , Mice , Mice, Transgenic , Molecular Docking Simulation , Orthomyxoviridae Infections/prevention & control , Swine , Swine Diseases/prevention & control , beta-Defensins/genetics , beta-Defensins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...