2.Clinical and drug sensitivity characteristics of invasive non-typhoidal Salmonella enteritis in children aged 0-6 years in Chengdu, China, 2022-2023.
Ling-Rong YANG ; Chun-Ting ZHOU ; Jing GUO ; Yu-Lu WU ; Fu XIONG
Chinese Journal of Contemporary Pediatrics 2025;27(3):315-320
OBJECTIVES:
To investigate the clinical characteristics and drug resistance profile of invasive non-typhoidal Salmonella (NTS) enteritis in children in Chengdu, China, providing a reference for rational drug use and empirical treatment in clinical practice.
METHODS:
A retrospective analysis was conducted on the clinical data of 130 children with invasive bacterial enteritis due to NTS identified by fecal bacterial culture and the results of drug sensitivity tests for NTS in Chengdu from January 2022 to December 2023.
RESULTS:
NTS infections were mainly observed from April to September (113 cases, 86.9%), with a peak in August (36 cases, 27.7%). Children aged <36 months accounted for 86.2% (112/130) of all cases, and the main symptoms were diarrhea (130 cases, 100%), fever (123 cases, 94.6%), and hematochezia (112 cases, 86.2%). The 130 NTS isolates exhibited a sensitivity rate of 64.6% to ceftriaxone and 63.8% to cefotaxime, and a sensitivity rate of >90.0% to piperacillin-tazobactam and nitrofurantoin (nitrofurans). The detection rate of multidrug-resistant strains was 48.5% (63/130), and the clinical efficacy of third-generation cephalosporins used in 38 patients (29.2%) was inconsistent with the results of drug sensitivity tests.
CONCLUSIONS
The peak of invasive NTS enteritis in children aged 0-6 years occurs in August in the Chengdu area, with a relatively high incidence rate in children aged <36 months. The situation of drug resistance is severe for NTS, and piperacillin-tazobactam may be an effective option for treating multidrug-resistant NTS infections in children, while nitrofuran antibiotics might be used to treat such infections.
Humans
;
Infant
;
Child, Preschool
;
Enteritis/microbiology*
;
Retrospective Studies
;
Male
;
Salmonella Infections/microbiology*
;
Female
;
Child
;
Salmonella/drug effects*
;
Infant, Newborn
;
Microbial Sensitivity Tests
;
Anti-Bacterial Agents/therapeutic use*
3.Clinical and Laboratory Characteristics of Streptococcus mitis Causing Bloodstream Infection in Children with Hematological Disease.
Yu-Long FAN ; Guo-Qing ZHU ; Zhi-Ying TIAN ; Yan-Xia LYU ; Zhao WANG ; Ye GUO ; Wen-Yu YANG ; Qing-Song LIN ; Xiao-Juan CHEN
Journal of Experimental Hematology 2025;33(1):286-291
OBJECTIVE:
To investigate the risk factors, clinical characteristics, and bacterial resistance of bloodstream infections caused by Streptococcus mitis in children with hematological disease, so as to provide a reference for infection control.
METHODS:
The clinical information and laboratory findings of pediatric patients complicated with blood cultures positive for Streptococcus mitis from January 2018 to December 2020 in the Institute of Hematology & Blood Diseases Hospital were searched and collected. The clinical characteristics, susceptibility factors, and antibiotic resistance of the children were retrospectively analyzed.
RESULTS:
Data analysis from 2018 to 2020 showed that the proportion of Streptococcus mitis isolated from bloodstream infections in children (≤14 years old) with hematological diseases was the highest (19.91%) and significantly higher than other bacteria, accounting for 38.64% of Gram-positive cocci, and presented as an increasing trend year by year. A total of 427 children tested positive blood cultures, including 85 children with bloodstream infections caused by Streptococcus mitis who tested after fever. Most children experienced a recurrent high fever in the early and middle stages (≤6 d) of neutropenia and persistent fever for more than 3 days. After adjusting the antibiotics according to the preliminary drug susceptibility results, the body temperature of most children (63.5%) returned to normal within 4 days. The 85 children were mainly diagnosed with acute myeloid leukemia (AML), accounting for 84.7%. The proportion of children in the neutropenia stage was 97.7%. The incidence of oral mucosal damage, lung infection, and gastrointestinal injury symptoms was 40%, 31.8%, and 27.1%, respectively. The ratio of elevated C-reactive protein (CRP) and procalcitonin was 65.9% and 9.4%, respectively. All isolated strains of Streptococcus mitis were not resistant to vancomycin and linezolid, and the resistance rate to penicillin, cefotaxime, levofloxacin, and quinupristin-dalfopristin was 10.6%, 8.2%, 9.4%, and 14.1%, respectively. None of children died due to bloodstream infection caused by Streptococcus mitis.
CONCLUSION
The infection rate of Streptococcus mitis is increasing year by year in children with hematological diseases, especially in children with AML. Among them, neutropenia and oral mucosal damage after chemotherapy are high-risk infection factors. The common clinical symptoms include persistent high fever, oral mucosal damage, and elevated CRP. Penicillin and cephalosporins have good sensitivity. Linezolid, as a highly sensitive antibiotic, can effectively control infection and shorten the course of disease.
Humans
;
Child
;
Streptococcal Infections/microbiology*
;
Retrospective Studies
;
Hematologic Diseases/complications*
;
Streptococcus mitis
;
Drug Resistance, Bacterial
;
Risk Factors
;
Microbial Sensitivity Tests
;
Anti-Bacterial Agents
;
Female
;
Male
;
Bacteremia/microbiology*
;
Child, Preschool
;
Adolescent
4.Autophagy reduces bacterial translocation by regulating intestinal mucosal oxidative stress.
Xing LU ; Chengfen YIN ; Yaxiao SU ; Xinjing GAO ; Fengmei WANG ; Lei XU
Chinese Critical Care Medicine 2025;37(2):153-159
OBJECTIVE:
To investigate the mechanism of autophagy in regulating bacterial translocation in intestinal infection caused by hypervirulent Klebsiella pneumonia (hvKp) and explore the method of reducing translocation infection of intestinal bacteria.
METHODS:
Fifty C57BL/6J mice were divided into gavage group (n = 40) and control group (CO group, n = 10). The gavage group was orally administered with 200 μL/d of hvKp (colony count of 109 CFU/mL) continuously for 5 days to establish a hvKp intestinal infection model. CO group was given an equal amount of normal saline. After the experiment, the mice were anesthetized with lsofluraneand euthanized with cervical dislocation under anesthesia. Peripheral venous blood of mice was collected to detect bacterial translocation by 16S rDNA sequencing, then divided into translocation group (BT+ group) and non-translocation group (BT- group). Hematoxylin-eosin (HE) staining was used to evaluate intestinal morphology. The ultrastructural changes of intestinal tissues were observed by electron microscope. The levels of intestinal oxidative stress indicators such as superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GPx) were measured. Translocation was detected by in situ hybridization. The expression of tight junction protein microtubule-associated protein 1 light chain 3-II (LC3-II) and autophagy protein Beclin-1 were measured by Western blotting. The mRNA expression of tight junction proteins ZO-1 and Claudin-2 were detected by reverse transcription-polymerase chain reaction (RT-PCR). The expression of autophagy protein and tight junction protein were observed by immunofluorescence.
RESULTS:
Two out of 40 mice in the gavage group died after developing aspiration pneumonia. All mice in the CO group survived. The 16S rDNA sequencing results showed that no bacteria were detected in the peripheral blood of the CO group, but bacteria were detected in the peripheral blood of 18 mice in the gavage group, with a bacterial translocation rate of 47.4%. The BT- and BT+ groups showed intestinal mucosal tissue damage, with severe damage in the BT+ group. Compared with the CO group, the level of MDA in the BT- and BT+ groups were significantly increased, while the activities of SOD and GPx were significantly decreased. Compared with the BT- group, the MDA level in the BT+ group further increased, while the SOD and GPx activities further decreased [MDA (mmol/mg): 2.98±0.11 vs. 2.48±0.11, SOD (U/mg): 62.40±5.45 vs. 73.40±4.08, GPx (U/mg): 254.72±10.80 vs. 303.55±8.57, all P < 0.01]. The results of in situ hybridization detection showed that after continuous gastric lavage for 5 days, displaced hvKp was detected in the intestinal mucosal lamina propria and liver tissue of the BT+ group. Compared with the CO group, the protein expressions of LC3-II and Beclin-1 in the BT- and BT+ groups were significantly increased. The protein expressions of LC3-II and Beclin-1 in the BT+ group were obviously lower than those in the BT- group (LC3-II/β-actin: 0.38±0.04 vs. 0.70±0.09, Beclin-1/β-actin: 0.62±0.05 vs. 0.86±0.05, both P < 0.01), and there were autophagosomes in the intestinal mucosa. These results indicated that intestinal mucosal autophagy was activated after hvKp continuous gavage. Compared with CO group, the mRNA expressions of ZO-1 and Claudin-2 in the BT- and BT+ groups were significantly decreased. Compared with the BT- group, the mRNA expressions of ZO-1 and Claudin-2 in the BT+ group was further reduced [ZO-1 mRNA (2-ΔΔCT): 0.78±0.06 vs. 0.88±0.06, Claudin-2 mRNA (2-ΔΔCT): 0.40±0.04 vs. 0.70±0.06, both P < 0.01]. The immunofluorescence results showed that the fluorescence intensity of LC3-II, Beclin-1, ZO-1, and Claudin-2 in the BT+ group was significantly lower than that in the BT- group.
CONCLUSION
HvKp can activate intestinal mucosal autophagy and reduce the damage to intestinal mucosal barrier function by down-regulating oxidative stress level, reduce the occurrence of bacterial translocation.
Animals
;
Oxidative Stress
;
Mice, Inbred C57BL
;
Autophagy
;
Intestinal Mucosa/microbiology*
;
Bacterial Translocation
;
Mice
;
Klebsiella Infections/microbiology*
;
Superoxide Dismutase/metabolism*
;
Beclin-1
5.Comprehensive analysis of the antibacterial activity of 5,8-dihydroxy-1,4-naphthoquinone derivatives against methicillin-resistant Staphylococcus aureus.
Qingqing CHEN ; Yuhang DING ; Zhongyi LI ; Xingyu CHEN ; Aliya FAZAL ; Yahan ZHANG ; Yudi MA ; Changyi WANG ; Liu YANG ; Tongming YIN ; Guihua LU ; Hongyan LIN ; Zhongling WEN ; Jinliang QI ; Hongwei HAN ; Yonghua YANG
Chinese Journal of Natural Medicines (English Ed.) 2025;23(5):604-613
Given the increasing concern regarding antibacterial resistance, the antimicrobial properties of naphthoquinones have recently attracted significant attention. While 1,4-naphthoquinone and its derivatives have been extensively studied, the antibacterial properties of 5,8-dihydroxy-1,4-naphthoquinone derivatives remain relatively unexplored. This study presents a comprehensive in vitro and in vivo analysis of the antibacterial activity of 35 naturally sourced and chemically synthesized derivatives of 5,8-dihydroxy-1,4-naphthoquinone. Kirby-Bauer antibiotic testing identified three compounds with activity against methicillin-resistant Staphylococcus aureus (MRSA), with one compound (PNP-02) demonstrating activity comparable to vancomycin in minimum inhibitory concentration, minimum bactericidal concentration (MBC), and time-kill assays. Microscopic and biochemical analyses revealed that PNP-02 adversely affects the cell wall and cell membrane of MRSA. Mechanistic investigations, including proteomic sequencing analyses, Western blotting, and RT-qPCR assays, indicated that PNP-02 compromises cell membrane integrity by inhibiting arginine biosynthesis and pyrimidine metabolism pathways, thereby increasing membrane permeability and inducing bacterial death. In an in vivo mouse model of skin wound healing, PNP-02 exhibited antibacterial efficacy similar to vancomycin. The compound demonstrated low toxicity to cultured human cells and in hemolysis assays and remained stable during serum incubation. These findings suggest that PNP-02 possesses promising bioactivity against MRSA and represents a potential novel antibacterial agent.
Methicillin-Resistant Staphylococcus aureus/genetics*
;
Anti-Bacterial Agents/chemistry*
;
Naphthoquinones/administration & dosage*
;
Animals
;
Microbial Sensitivity Tests
;
Mice
;
Humans
;
Staphylococcal Infections/microbiology*
;
Molecular Structure
6.Deciphering the therapeutic potential and mechanisms of Artemisia argyit essential oil on flagellum-mediated Salmonella infections.
Linlin DING ; Lei XU ; Na HU ; Jianfeng WANG ; Jiazhang QIU ; Qingjie LI ; Xuming DENG
Chinese Journal of Natural Medicines (English Ed.) 2025;23(6):714-726
Salmonellosis represents a global epidemic, and the emergence of extensively drug-resistant (XDR) Salmonella and its sustained transmission worldwide constitutes a significant public health concern. Flagellum-mediated motility serves as a crucial virulence trait of Salmonella that guides the pathogen toward the epithelial surface, enhancing gut colonization. Artemisia argyit essential oil, a traditional herb extract, demonstrates efficacy in treating inflammation-related symptoms and diseases; however, its effects on flagellum assembly and expression mechanisms in anti-Salmonella activity remain inadequately explored. This study aimed to elucidate the mechanism by which Artemisia argyit essential oil addresses Salmonella infections. Network pharmacological analysis revealed that Traditional Chinese Medicine (TCM) Artemisia argyit exhibited anti-Salmonella infection potential and inhibited flagellum-dependent motility. The application of Artemisia argyit essential oil induced notable motility defects through the downregulation of flagellar and fimbriae expression. Moreover, it significantly reduced Salmonella-infected cell damage by interfering with flagellum-mediated Salmonella colonization. In vivo studies demonstrated that Artemisia argyit essential oil administration effectively alleviated Salmonella infection symptoms by reducing bacterial loads, inhibiting interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) production, and diminishing pathological injury. Gas chromatography-mass spectrometry (GC-MS) analysis identified forty-three compounds in Artemisia argyit essential oil, with their corresponding targets and active ingredients predicted. Investigation of an in vivo model of Salmonella infection using the active ingredient demonstrated that alpha-cedrene ameliorated Salmonella infection. These findings suggest the potential application of Artemisia argyit essential oil in controlling Salmonella, the predominant food-borne pathogen.
Artemisia/chemistry*
;
Oils, Volatile/chemistry*
;
Animals
;
Flagella/drug effects*
;
Salmonella Infections/microbiology*
;
Humans
;
Mice
;
Anti-Bacterial Agents/pharmacology*
;
Salmonella/pathogenicity*
7.Multidrug resistance of Helicobacter pylori and its impact on the diagnosis and treatment of gastrointestinal diseases and countermeasures.
Xiya YAN ; Canlin ZHENG ; Zhihui TANG ; Youjun FENG ; Baoning WANG
Chinese Journal of Biotechnology 2025;41(4):1240-1251
Helicobacter pylori is a bacterium that can cause chronic gastritis, peptic ulcers, and other gastrointestinal diseases. The World Health Organization has classified H. pylori as a group Ⅰ carcinogen. Antibiotics are the primary clinical approach for eradicating H. pylori. However, incomplete eradication of H. pylori by antibiotics can lead to persistent infection, which is a major risk factor for the high incidence of gastric cancer. The widespread use of antibiotics has led to the emergence of multidrug resistance in H. pylori, contributing to treatment failures of chronic gastric diseases and increasing the risk of spreading resistant strains. Multidrug-resistant H. pylori has become a serious challenge in the diagnosis and treatment of gastrointestinal diseases. This paper reviews the global trends in the development of multidrug resistance in H. pylori, the underlying mechanisms, the challenges it poses to clinical diagnosis, and its impact on drug development, drawing on relevant literature and the research findings from our group. It proposes using cgt expression as a novel method for determining viable bacteria, identifying intracellularization as a new form of resistance in H. pylori, and exploring the potential of O-glycans as a therapeutic approach against H. pylori to address multidrug resistance. It provides new insights into understanding the mechanisms of H. pylori multidrug resistance and its prevention strategies, offering promising directions for future clinical treatments and antimicrobial drug development.
Helicobacter pylori/genetics*
;
Humans
;
Drug Resistance, Multiple, Bacterial
;
Helicobacter Infections/microbiology*
;
Anti-Bacterial Agents/therapeutic use*
;
Gastrointestinal Diseases/drug therapy*
8.Phage therapy for multidrug-resistant Acinetobacter baumannii.
Chinese Journal of Biotechnology 2025;41(6):2256-2274
Acinetobacter baumannii is a Gram-negative opportunistic pathogen widely distributed in hospital settings. It can survive for a long time and cause a variety of infections, including pneumonia, septicemia, urinary tract infections, and meningitis. The bacterium demonstrates extensive resistance, particularly to critical antibiotics like carbapenems and polymyxins, posing a serious threat to the recovery of severely ill patients. Carbapenem-resistant A. baumannii has been designated as a pathogen of critical priority on the World Health Organization (WHO) Bacterial Pathogen Priority List, requiring urgent development of new therapeutic agents. Phages, as a novel biological control approach, exhibit substantial potential in combating A. baumannii infections due to their specific ability to infect and lyse bacteria. This review highlights the application and potential of phages and phage-derived enzymes against multidrug-resistant A. baumannii, considering the epidemiological trends of A. baumannii in China, with the aim of providing innovative insights and strategies for phage therapy of drug-resistant bacterial infections.
Acinetobacter baumannii/drug effects*
;
Drug Resistance, Multiple, Bacterial
;
Phage Therapy/methods*
;
Acinetobacter Infections/microbiology*
;
Humans
;
Bacteriophages/physiology*
;
Anti-Bacterial Agents/pharmacology*
9.Impacts of Helicobacter pylori infection and eradication on gastrointestinal microbiota: An up-to-date critical review and future perspectives.
Yu LI ; Cong HE ; Nonghua LU
Chinese Medical Journal 2024;137(23):2833-2842
Helicobacter pylori ( H. pylori ) infects approximately half of the population worldwide and causes chronic gastritis, peptic ulcers, and gastric cancer. Test-and-treat strategies have been recommended for the prevention of H. pylori -associated diseases. Advancements in high-throughput sequencing technologies have broadened our understanding of the complex gastrointestinal (GI) microbiota and its role in maintaining host homeostasis. Recently, an increasing number of studies have indicated that the colonization of H. pylori induces dramatic alterations in the gastric microbiota, with a predominance of H. pylori and a reduction in microbial diversity. Dysbiosis of the gut microbiome has also been observed after H. pylori infection, which may play a role in the development of colorectal cancer. However, there is concern regarding the impact of antibiotics on the gut microbiota during H. pylori eradication. In this review, we summarize the current literature concerning how H. pylori infection reshapes the GI microbiota and the underlying mechanisms, including changes in the gastric environment, immune responses, and persistent inflammation. Additionally, the impacts of H. pylori eradication on GI microbial homeostasis and the use of probiotics as adjuvant therapy are also discussed. The shifts in the GI microbiota and their crosstalk with H. pylori may provide potential targets for H. pylori -related gastric diseases and extragastric manifestations.
Helicobacter Infections/microbiology*
;
Humans
;
Helicobacter pylori/pathogenicity*
;
Gastrointestinal Microbiome/drug effects*
;
Probiotics/therapeutic use*
;
Anti-Bacterial Agents/therapeutic use*
10.A single-center retrospective study of pathogen distribution and antibiotic resistance of bloodstream infections in emergency department.
Yishu TANG ; Lihua CHEN ; Jie XIAO ; Kun YAN ; Jing QI ; Kefu ZHOU ; Huaizheng LIU
Journal of Central South University(Medical Sciences) 2024;49(11):1799-1807
OBJECTIVES:
Bloodstream infections in emergency patients have a high incidence, severe disease progression, and rapid deterioration. Early administration of appropriate antimicrobial agents is crucial for improving patient outcomes. This study aims to investigate the incidence, pathogen distribution, and antimicrobial resistance patterns of bloodstream infections in emergency patients, providing a reference for rational antibiotic use in clinical practice.
METHODS:
Medical records of patients diagnosed with bloodstream infections in the emergency department of a hospital in Hunan Province between January 2018 and October 2022 were retrospectively collected. Clinical characteristics of bloodstream infection patients were analyzed, and the distribution trends and antimicrobial susceptibility of clinical isolates were examined.
RESULTS:
During the study period, 2 215 blood culture samples were submitted from the emergency department, with a positivity rate of 13.27%. After excluding eight cases with missing data or suspected contamination, 286 patients with bloodstream infections were included, with community-acquired infections accounting for the majority (85.66%). The most common primary infection site was the urinary tract (24.48%), followed by respiratory tract infections (20.28%) and biliary and intra-abdominal infections (17.13%). The 30-day mortality rate of bloodstream infections was 16.08%. A total of 286 pathogens were isolated, including 181 (63.29%) Gram-negative bacteria, primarily Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa; 101 (35.31%) Gram-positive bacteria, mainly Staphylococcus aureus, Staphylococcus epidermidis, and Streptococcus pneumoniae; and only 4 (1.40%) fungal isolates. Antimicrobial susceptibility testing showed that the key Enterobacteriaceae strains exhibited resistance rates of 2.4% to carbapenems, 16.3% to piperacillin sodium and tazobactam sodium, and 15.3% to ceftazidime, with no detected resistance to tigecycline or polymyxins. The main non-fermentative bacteria showed resistance rates of 29.6% to piperacillin sodium and tazobactam sodium, 13.3% to cefoperazone sodium and sulbactam sodium, and 27.1% to quinolones. Among Gram-negative bacteria, multidrug-resistant strains accounted for 40.9% (74/181), with carbapenem-resistant Escherichia coli and Klebsiella pneumoniae detected in 5.4% (5/92) and 13.6% (6/44) of cases, respectively. No carbapenem-resistant Pseudomonas aeruginosa was identified. Among Gram-positive bacteria, resistance rates to penicillin G, rifampicin, and cefoxitin were 74.7%, 4.2%, and 50%, respectively, with only 3 cases of resistant to glycopeptide antibiotics.
CONCLUSIONS
Bloodstream infections in emergency patients are predominantly community-acquired, with Gram-negative bacteria being the most common pathogens. The isolated pathogens exhibited relatively low resistance rates to commonly used clinical antibiotics.
Retrospective Studies
;
Emergency Service, Hospital/statistics & numerical data*
;
Drug Resistance, Bacterial
;
Anti-Bacterial Agents/therapeutic use*
;
Incidence
;
Microbial Sensitivity Tests/statistics & numerical data*
;
Bacteremia/microbiology*
;
Community-Acquired Infections/microbiology*
;
Gram-Negative Bacteria/isolation & purification*
;
Blood Culture/statistics & numerical data*
;
Humans
;
Male
;
Female
;
Adolescent
;
Young Adult
;
Adult
;
Middle Aged
;
Aged
;
Aged, 80 and over
;
China/epidemiology*

Result Analysis
Print
Save
E-mail