1
|
Campos-Madueno EI, Aldeia C, Roumet MC, Limacher A, Sendi P, Endimiani A. Epidemiology and risk factors of expatriates returning to Switzerland colonized at the intestinal level with multidrug-resistant Enterobacterales. Eur J Clin Microbiol Infect Dis 2025; 44:1007-1014. [PMID: 39953366 PMCID: PMC11947073 DOI: 10.1007/s10096-025-05069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Living in high-endemic regions increases the risk of intestinal colonization by multidrug-resistant Enterobacterales (MDR-Ent). This study investigated Swiss expatriates residing abroad (≥ 3 months) to assess their colonization status upon returning to Switzerland. Selective culture-based methods were implemented to detect third-generation cephalosporins- (3GC-R), carbapenems- (CR), and colistin-resistant (COL-R) strains. Whole-genome sequencing was used to characterize antimicrobial resistance genes, sequence type (ST), and phylogroup of MDR-Ent. Epidemiological data were analyzed using uni- and multivariable models to identify risk factors, providing crude and adjusted odds ratios (ORs). Among 196 participants living across Africa, Asia, the Americas, and Europe, the overall MDR-Ent colonization prevalence was 42.9%. Continent of residence emerged as a significant risk factor (p = 0.04) for colonization: Africa (adjusted OR = 3.4, 95% CI 1.0-11.0) and Asia (adjusted OR = 4.7, 95% CI 1.5-15.0). Extended-spectrum β-lactamase-producing Escherichia coli (Ec) was the most frequent isolated species (n = 107 out of 119 Ent). Most 3GC-R-Ec possessed blaCTX-M genes (n = 89; 83.2%) and pandemic lineages were frequent (e.g., ST69 and ST131, n = 18). No CR-Ent were detected, but some COL-R strains (n = 18; of which 15 Ec) harbored the mcr-1.1 gene. Expatriates represent an understudied population at risk of MDR-Ent colonization. This population may contribute to the importation and potential dissemination of dangerous bacteria into low-prevalence countries, as shown in this Swiss study, warranting further investigation and surveillance.
Collapse
Affiliation(s)
- Edgar I Campos-Madueno
- Institute for Infectious Diseases (IFIK), University of Bern, Friedbühlstrasse 25, Bern, CH-3001, Switzerland
| | - Claudia Aldeia
- Institute for Infectious Diseases (IFIK), University of Bern, Friedbühlstrasse 25, Bern, CH-3001, Switzerland
| | - Marie C Roumet
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Andreas Limacher
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Functioning Information Reference Lab, Swiss Paraplegic Research, Nottwil, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases (IFIK), University of Bern, Friedbühlstrasse 25, Bern, CH-3001, Switzerland
| | - Andrea Endimiani
- Institute for Infectious Diseases (IFIK), University of Bern, Friedbühlstrasse 25, Bern, CH-3001, Switzerland.
| |
Collapse
|
2
|
Rivera-Rodriguez DE, Busby C, Cervantes-Barragan L, Weiss DS. Widespread heteroresistance to antibiotics in Lactobacillus species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644958. [PMID: 40196655 PMCID: PMC11974758 DOI: 10.1101/2025.03.24.644958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Lactobacilli are prevalent members of the intestinal and reproductive tract microbiota of humans and other species. They are commonly used in probiotics and various food products due to their beneficial effects on human health. For example, these beneficial microbes are used to treat diarrhea caused by antibiotic therapy and are commonly given during antibiotic treatment. Despite the many studies conducted to understand the beneficial effects of Lactobacilli, less is known about their resistance and heteroresistance to antibiotics. In this study, we evaluated the resistance heterogeneity in eight Lactobacillus species. Our results demonstrate that several Lactobacilli species, including Lactobacillus rhamnosus , are heteroresistant to antibiotics, a recently discovered phenotype commonly seen in multidrug-resistant organisms that cause clinical failures but understudied in commensals and probiotics.
Collapse
|
3
|
Ture Z, İskender G, Serhat Şahinoğlu M, Beste Özkara E, Kalem AK, Eryılmaz Eren E, Ürkmez FY, Çetin S, Azak E, Erdem İ, Rello J, Alp E. Epidemiology and prognosis of sepsis in cancer patients: A multicenter prospective observational study. Am J Med Sci 2025:S0002-9629(25)00923-1. [PMID: 39952291 DOI: 10.1016/j.amjms.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/16/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVES To determine the epidemiology and prognosis of sepsis in cancer patients and the influence of sepsis on the mortality of cancer patients. METHODS In this multicenter, prospective, observational study, cancer patients hospitalized without sepsis followed up until discharge or a maximum of 90 days were included. The incidence of sepsis in the follow-up period, risk factors for sepsis, risk factors for 28-day mortality in patients with sepsis and 90-day mortality in the entire group were determined. RESULTS During the study, 790 cancer patients were included. Sepsis developed in 72 patients (9.1 %) during the follow-up. Older age, hospitalization due to any infection, graft versus host diseases (GVHD), the presence of a urethral catheter, and previous bacterial infection in the last three months were risk factors of sepsis. Among all cancer patients, sepsis was found to be the most important factor influencing 90-day mortality (OR 13.42(1.79-6.83)). Mortality among the sepsis cohort was independently associated with an infection with a carbapenem-resistant bacterium (OR 15.47(1.45-64.17)), appropriateness of empirical treatment (OR 5.02 (0.17-7.61) and having a clinical improvement on the fifth day of the treatment (OR 10.58(0.39-28.25). CONCLUSIONS Sepsis was documented in one out of 11 hospitalized cancer patients and the mortality rate increases 13-fold when sepsis develops. Invasive devices, GVHD, and previous bacterial infections were related to sepsis and antibiotic resistance were the most important driver for mortality. Antimicrobial stewardship, rational use of catheters (if necessary, in accordance with asepsis/antisepsis, short-term use) is important to save lives in cancer patients.
Collapse
Affiliation(s)
- Zeynep Ture
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, , Türkiye.
| | - Gülşen İskender
- University of Health Sciences Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital. Infectious Diseases and Clinical Microbiology Clinic, Ankara, Türkiye
| | | | - Ezel Beste Özkara
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Celal Bayar University, Manisa, Türkiye
| | - Ayşe Kaya Kalem
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara City Hospital, Ankara, Türkiye
| | - Esma Eryılmaz Eren
- Department of Infectious Disease and Clinical Microbiology, Kayseri City Trainig and Research Hospital, University of Health Sciences, Kayseri, Turkiye
| | - Fatma Yekta Ürkmez
- Kırıkkale Yüksek İhtisas Hospital, Clinic of Infectious Diseases and Clinical Microbiology, Kırıkkale, Türkiye
| | - Sinan Çetin
- Department of Infectious Diseases and Clinical Microbiology, Giresun University, Giresun, Turkiye
| | - Emel Azak
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Kocaeli University, Kocaeli, Türkiye
| | - İlknur Erdem
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Namik Kemal University, Tekirdag, Turkiye
| | - Jordi Rello
- Vall d'Hebron Research Institute, Pneumonia and Clinical Research/Epidemiology Sepsis (CRIPSES) Unit, VHIR & CIBER, Madrid, Spain
| | - Emine Alp
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara City Hospital, Ankara, Türkiye
| |
Collapse
|
4
|
Mafe AN, Büsselberg D. Microbiome Integrity Enhances the Efficacy and Safety of Anticancer Drug. Biomedicines 2025; 13:422. [PMID: 40002835 PMCID: PMC11852609 DOI: 10.3390/biomedicines13020422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The intricate relationship between anticancer drugs and the gut microbiome influences cancer treatment outcomes. This review paper focuses on the role of microbiome integrity in enhancing the efficacy and safety of anticancer drug therapy, emphasizing the pharmacokinetic interactions between anticancer drugs and the gut microbiota. It explores how disruptions to microbiome composition, or dysbiosis, can alter drug metabolism, immune responses, and treatment side effects. By examining the mechanisms of microbiome disruption caused by anticancer drugs, this paper highlights specific case studies of drugs like cyclophosphamide, 5-fluorouracil, and irinotecan, and their impact on microbial diversity and clinical outcomes. The review also discusses microbiome-targeted strategies, including prebiotics, probiotics, postbiotics, and fecal microbiota transplantation (FMT), as promising interventions to enhance cancer treatment. Furthermore, the potential of microbiome profiling in personalizing therapy and integrating these interventions into clinical practice is explored. Finally, this paper proposes future research directions, including developing novel biomarkers and a deeper comprehension of drug-microbiome interactions, to respond to current gaps in knowledge and improve patient outcomes in cancer care.
Collapse
Affiliation(s)
- Alice N. Mafe
- Department of Biological Sciences, Faculty of Sciences, Taraba State University, Main Campus, Jalingo 660101, Taraba State, Nigeria;
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha Metropolitan Area, Doha P.O. Box 22104, Qatar
| |
Collapse
|
5
|
Li S, Li R, Lee JR, Zhao N, Ling W. ZINQ-L: a zero-inflated quantile approach for differential abundance analysis of longitudinal microbiome data. Front Genet 2025; 15:1494401. [PMID: 39944355 PMCID: PMC11814158 DOI: 10.3389/fgene.2024.1494401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025] Open
Abstract
Background Identifying bacterial taxa associated with disease phenotypes or clinical treatments over time is critical for understanding the underlying biological mechanism. Association testing for microbiome data is already challenging due to its complex distribution that involves sparsity, over-dispersion, heavy tails, etc. The longitudinal nature of the data adds another layer of complexity - one needs to account for the within-subject correlations to avoid biased results. Existing longitudinal differential abundance approaches usually depend on strong parametric assumptions, such as zero-inflated normal or negative binomial. However, the complex microbiome data frequently violate these distributional assumptions, leading to inflated false discovery rates. In addition, the existing methods are mostly mean-based, unable to identify heterogeneous associations such as tail events or subgroup effects, which could be important biomedical signals. Methods We propose a zero-inflated quantile approach for longitudinal (ZINQ-L) microbiome differential abundance test. A mixed-effects quantile rank-score-based test was proposed for hypothesis testing, which consists of a test in mixed-effects logistic model for the presence-absence status of the investigated taxon, and a series of mixed-effects quantile rank-score tests adjusted for zero inflation given its presence. As a regression method with minimal distributional assumptions, it is robust to the complex microbiome data, controlling false discovery rate, and is flexible to adjust for important covariates. Its comprehensive examination of the abundance distribution enables the identification of heterogeneous associations, improving the testing power. Results Extensive simulation studies and an application to a real kidney transplant microbiome study demonstrate the improved power of ZINQ-L in detecting true signals while controlling false discovery rates. Conclusion ZINQ-L is a zero-inflated quantile-based approach for detecting individual taxa associated with outcomes or exposures in longitudinal microbiome studies, providing a robust and powerful option to improve and complement the existing methods in the field.
Collapse
Affiliation(s)
- Shuai Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Runzhe Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - John R. Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Medical College of Cornell University, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Wodan Ling
- Division of Biostatistics, Department of Population Health Sciences, Weill Medical College of Cornell University, New York, NY, United States
| |
Collapse
|
6
|
Varela-Nájera RG, De la Cruz MA, Soria-Bustos J, González-Horta C, Delgado-Gardea MCE, Yáñez-Santos JA, Cedillo ML, Hirakawa H, Fox JG, Sánchez-Ramírez B, Ares MA. The Response Regulator OmpR Negatively Controls the Expression of Genes Implicated in Tilimycin and Tilivalline Cytotoxin Production in Klebsiella oxytoca. Microorganisms 2025; 13:158. [PMID: 39858926 PMCID: PMC11767513 DOI: 10.3390/microorganisms13010158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Klebsiella oxytoca toxigenic strains represent a critical health threat, mainly due to their link to antibiotic-associated hemorrhagic colitis. This serious condition results from the bacteria's ability to produce tilimycin and tilivalline cytotoxins. Our research highlights the pivotal role of OmpR, a key regulator within the EnvZ/OmpR two-component system, in controlling the virulence factors associated with K. oxytoca. Our findings strongly indicate that OmpR is a repressor of the aroX and npsA genes, the first genes of aroX and NRPS operons, respectively, which are indispensable for producing these enterotoxins. Notably, in the absence of OmpR, we observe a significant increase in cytotoxic effects on Caco-2 cells. These observations identify OmpR as a crucial negative transcription regulator for both operons, effectively managing the release of these cytotoxins. This research deepens our understanding of the mechanisms of toxigenic K. oxytoca and opens promising avenues for targeting OmpR for new therapeutic interventions. By focusing on this innovative approach, we can develop more effective solutions to combat this pressing health challenge, ultimately improving patient outcomes against this pathogen.
Collapse
Affiliation(s)
- Ramón G. Varela-Nájera
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua 31125, Mexico; (C.G.-H.); (M.C.E.D.-G.); (B.S.-R.)
| | - Miguel A. De la Cruz
- Centro de detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (M.A.D.l.C.); (J.S.-B.); (J.A.Y.-S.); (M.L.C.)
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Puebla 72410, Mexico
| | - Jorge Soria-Bustos
- Centro de detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (M.A.D.l.C.); (J.S.-B.); (J.A.Y.-S.); (M.L.C.)
| | - Carmen González-Horta
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua 31125, Mexico; (C.G.-H.); (M.C.E.D.-G.); (B.S.-R.)
| | - Ma Carmen E. Delgado-Gardea
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua 31125, Mexico; (C.G.-H.); (M.C.E.D.-G.); (B.S.-R.)
| | - Jorge A. Yáñez-Santos
- Centro de detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (M.A.D.l.C.); (J.S.-B.); (J.A.Y.-S.); (M.L.C.)
| | - María L. Cedillo
- Centro de detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (M.A.D.l.C.); (J.S.-B.); (J.A.Y.-S.); (M.L.C.)
| | - Hidetada Hirakawa
- Department of Bacteriology, Graduate School of Medicine, Gunma University, Maebashi 371-8514, Japan;
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Blanca Sánchez-Ramírez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Circuito Universitario Campus II, Chihuahua 31125, Mexico; (C.G.-H.); (M.C.E.D.-G.); (B.S.-R.)
| | - Miguel A. Ares
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| |
Collapse
|
7
|
Tahmasebi H, Arjmand N, Monemi M, Babaeizad A, Alibabaei F, Alibabaei N, Bahar A, Oksenych V, Eslami M. From Cure to Crisis: Understanding the Evolution of Antibiotic-Resistant Bacteria in Human Microbiota. Biomolecules 2025; 15:93. [PMID: 39858487 PMCID: PMC11764262 DOI: 10.3390/biom15010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The growing prevalence of antibiotic-resistant bacteria within the human microbiome has become a pressing global health crisis. While antibiotics have revolutionized medicine by significantly reducing mortality and enabling advanced medical interventions, their misuse and overuse have led to the emergence of resistant bacterial strains. Key resistance mechanisms include genetic mutations, horizontal gene transfer, and biofilm formation, with the human microbiota acting as a reservoir for antibiotic resistance genes (ARGs). Industrialization and environmental factors have exacerbated this issue, contributing to a rise in infections with multidrug-resistant (MDR) bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA) and carbapenem-resistant Enterobacteriaceae. These resistant pathogens compromise the effectiveness of essential treatments like surgical prophylaxis and chemotherapy, increase healthcare costs, and prolong hospital stays. This crisis highlights the need for a global One-Health approach, particularly in regions with weak regulatory frameworks. Innovative strategies, including next-generation sequencing (NGS) technologies, offer promising avenues for mitigating resistance. Addressing this challenge requires coordinated efforts, encompassing research, policymaking, public education, and antibiotic stewardship, to safeguard current antibiotics and foster the development of new therapeutic solutions. An integrated, multidimensional strategy is essential to tackle this escalating problem and ensure the sustainability of effective antimicrobial treatments.
Collapse
Affiliation(s)
- Hamed Tahmasebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud 36147-73943, Iran
| | - Neda Arjmand
- Department of Obstetrics and Gynecology, Tehran Medical University, Tehran 14167-53955, Iran
| | - Marzieh Monemi
- Department of Basic Science, Faculty of Pharmacy and Pharmaceutical Science, Tehran Medical Science, Islamic Azad University, Tehran 19395-1495, Iran
| | - Ali Babaeizad
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Farnaz Alibabaei
- Student Research Committee, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Negar Alibabaei
- Student Research Committee, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Aisa Bahar
- Department of Biochemistry, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | | | - Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
- Department of Bacteriology and Virology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| |
Collapse
|
8
|
Litvinov E, Litvinov A. Impact of Clindamycin on the Oral-Gut Axis: Gastrointestinal Side Effects and Clostridium difficile Infection in 45 Patients. Cureus 2024; 16:e75381. [PMID: 39781176 PMCID: PMC11710861 DOI: 10.7759/cureus.75381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/12/2025] Open
Abstract
Introduction The use of antibiotics such as oral clindamycin has been effective in treating bacterial infections. However, this medication often comes with significant side effects, particularly those affecting the gastrointestinal (GI) system. This study aims to evaluate the impact of different doses of clindamycin on GI health, specifically examining side effects like stomach upset, diarrhea duration, stomach pain, and recovery time. Given that clindamycin is frequently prescribed, understanding its impact on the oral-gut axis is critical to optimizing antibiotic therapy and reducing adverse events. Background Clindamycin, a lincosamide antibiotic, is widely used to treat a variety of bacterial infections. It acts by inhibiting bacterial protein synthesis but, like many antibiotics, also has unintended consequences for human gut health. The oral-gut axis represents a complex connection where antibiotics, such as clindamycin, can significantly alter the microbiota, leading to imbalances that manifest as diarrhea, abdominal pain, and other digestive issues. This study aims to explore these effects in depth by comparing two common doses of clindamycin, 300 mg versus 600 mg, and the impact of each dose on the severity and duration of GI side effects. Materials and methods This study involves 45 patients prescribed clindamycin for various bacterial infections. The patients were evaluated in two groups: 22 patients who received 300 mg and 23 patients who received 600 mg. Treatment duration ranged from seven to 10 days. Data collection focused on patient-reported symptoms, including the presence and duration of stomach upset, the length of diarrhea episodes, the persistence of stomach pain, and the overall recovery time. Statistical analysis included independent t-tests to compare symptom severity between the groups and chi-squared tests to assess differences in the incidence of side effects, while regression analysis was used to examine predictors of prolonged GI symptoms. Results The results of the study showed that 98% of patients experienced some side effects from oral clindamycin. Among those receiving the 600 mg dose, the frequency and severity of side effects were significantly higher compared to the 300 mg group. Specifically, the average duration of diarrhea in the 600 mg group was five days, compared to three days in the 300 mg group. Similarly, the average length of stomach pain in the higher dose group was seven days, compared to four days for those taking the lower dose. Chi-squared analysis indicated a significant association between the higher dose and increased incidence of GI symptoms. Regression analysis further showed that the 600 mg dose was a significant predictor of prolonged GI disturbances, underscoring a dose-dependent relationship. Conclusion The findings of this case study highlight that oral clindamycin, particularly at higher doses, is associated with increased GI side effects, including prolonged diarrhea and stomach pain. Almost all patients experienced side effects, with those on the 600 mg dose suffering more severe and prolonged symptoms compared to those on the 300 mg dose. The results suggest avoiding the prescription of oral clindamycin unless absolutely necessary, to reduce adverse outcomes and improve compliance. It is recommended to prioritize first-line antibiotics and reserve oral clindamycin as a secondary option. Further research is needed to investigate strategies for prescribing.
Collapse
Affiliation(s)
| | - Alan Litvinov
- Private Practice and Research, American Dental Association, Penfield, USA
| |
Collapse
|
9
|
Russo C, Surdo S, Valle MS, Malaguarnera L. The Gut Microbiota Involvement in the Panorama of Muscular Dystrophy Pathogenesis. Int J Mol Sci 2024; 25:11310. [PMID: 39457092 PMCID: PMC11508360 DOI: 10.3390/ijms252011310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Muscular dystrophies (MDs) are genetically heterogeneous diseases characterized by primary skeletal muscle atrophy. The collapse of muscle structure and irreversible degeneration of tissues promote the occurrence of comorbidities, including cardiomyopathy and respiratory failure. Mitochondrial dysfunction leads to inflammation, fibrosis, and adipogenic cellular infiltrates that exacerbate the symptomatology of MD patients. Gastrointestinal disorders and metabolic anomalies are common in MD patients and may be determined by the interaction between the intestine and its microbiota. Therefore, the gut-muscle axis is one of the actors involved in the spread of inflammatory signals to all muscles. In this review, we aim to examine in depth how intestinal dysbiosis can modulate the metabolic state, the immune response, and mitochondrial biogenesis in the course and progression of the most investigated MDs such as Duchenne Muscular Dystrophy (DMD) and Myotonic Dystrophy (MD1), to better identify gut microbiota metabolites working as therapeutic adjuvants to improve symptoms of MD.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
10
|
Gottwald J, Balke J, Stellmacher J, van Vorst K, Ghazisaeedi F, Fulde M, Alexiev U. Cy3-Based Nanoviscosity Determination of Mucus: Effect of Mucus Collection Methods and Antibiotics Treatment. Macromol Biosci 2024; 24:e2300437. [PMID: 38625085 DOI: 10.1002/mabi.202300437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/21/2023] [Indexed: 04/17/2024]
Abstract
The integrity of the protective mucus layer as a primary defense against pathogen invasion and microbial leakage into the intestinal epithelium can be compromised by the effects of antibiotics on the commensal microbiome. Changes in mucus integrity directly affect the solvent viscosity in the immediate vicinity of the mucin network, that is, the nanoviscosity, which in turn affects both biochemical reactions and selective transport. To assess mucus nanoviscosity, a reliable readout via the viscosity-dependent fluorescence lifetime of the molecular rotor dye cyanine 3 is established and nanoviscosities from porcine and murine ex vivo mucus are determined. To account for different mucin concentrations due to the removal of digestive residues during mucus collection, the power law dependence of mucin concentration on viscosity is used. The impact of antibiotics combinations (meropenem/vancomycin, gentamycin/ampicillin) on ex vivo intestinal mucus nanoviscosity is presented. The significant increase in viscosity of murine intestinal mucus after treatment suggests an effect of antibiotics on the microbiota that affects mucus integrity. This method will be a useful tool to assess how drugs, directly or indirectly, affect mucus integrity. Additionally, the method can be utilized to analyze the role of mucus nanoviscosity in health and disease, as well as in drug development.
Collapse
Affiliation(s)
- Jacqueline Gottwald
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Jens Balke
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Johannes Stellmacher
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| | - Kira van Vorst
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Robert-von-Ostertag-Str. 7, 14163, Berlin, Germany
| | - Ulrike Alexiev
- Physics Department, Freie Universität Berlin, Arnimallee 14, 14195, Berlin, Germany
| |
Collapse
|
11
|
Fu Y, Cheng HW. The Influence of Cecal Microbiota Transplantation on Chicken Injurious Behavior: Perspective in Human Neuropsychiatric Research. Biomolecules 2024; 14:1017. [PMID: 39199404 PMCID: PMC11352350 DOI: 10.3390/biom14081017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Numerous studies have evidenced that neuropsychiatric disorders (mental illness and emotional disturbances) with aggression (or violence) pose a significant challenge to public health and contribute to a substantial economic burden worldwide. Especially, social disorganization (or social inequality) associated with childhood adversity has long-lasting effects on mental health, increasing the risk of developing neuropsychiatric disorders. Intestinal bacteria, functionally as an endocrine organ and a second brain, release various immunomodulators and bioactive compounds directly or indirectly regulating a host's physiological and behavioral homeostasis. Under various social challenges, stress-induced dysbiosis increases gut permeability causes serial reactions: releasing neurotoxic compounds, leading to neuroinflammation and neuronal injury, and eventually neuropsychiatric disorders associated with aggressive, violent, or impulsive behavior in humans and various animals via a complex bidirectional communication of the microbiota-gut-brain (MGB) axis. The dysregulation of the MGB axis has also been recognized as one of the reasons for the prevalence of social stress-induced injurious behaviors (feather pecking, aggression, and cannibalistic pecking) in chickens. However, existing knowledge of preventing and treating these disorders in both humans and chickens is not well understood. In previous studies, we developed a non-mammal model in an abnormal behavioral investigation by rationalizing the effects of gut microbiota on injurious behaviors in chickens. Based on our earlier success, the perspective article outlines the possibility of reducing stress-induced injurious behaviors in chickens through modifying gut microbiota via cecal microbiota transplantation, with the potential for providing a biotherapeutic rationale for preventing injurious behaviors among individuals with mental disorders via restoring gut microbiota diversity and function.
Collapse
Affiliation(s)
- Yuechi Fu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Heng-Wei Cheng
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Lear L, Hesse E, Buckling A. Disturbances can facilitate prior invasions more than subsequent invasions in microbial communities. Ecol Lett 2024; 27:e14493. [PMID: 39140430 DOI: 10.1111/ele.14493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/02/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024]
Abstract
Invasions are commonly found to benefit from disturbance events. However, the importance of the relative timing of the invasion and disturbance for invader success and impact on community composition remains uncertain. Here, we experimentally test this by invading a five-species bacterial community on eight separate occasions-four before a disturbance and four after. Invader success and impact on community composition was greatest when the invasion immediately followed the disturbance. However, the subsequent invasions had negligible success or impact. Pre-disturbance, invader success and impact was greatest when the invader was added just before the disturbance. Importantly, however, the first three pre-disturbance invasion events had significantly greater success than the last three post-disturbance invasions. Moreover, these findings were consistent across a range of propagule pressures. Overall, we demonstrate that timing is highly important for both the success and impact on community composition of an invader, with both being lower as time since disturbance progresses.
Collapse
Affiliation(s)
- Luke Lear
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| | - Elze Hesse
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| | - Angus Buckling
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| |
Collapse
|
13
|
Tan X, Liu H, Qiu W, Li Z, Ge S, Luo Y, Zeng N, Chen M, Zhou Q, Cai S, Long J, Cen Z, Su J, Zhou H, He X. The nasal microbiota is a potential diagnostic biomarker for sepsis in critical care units. Microbiol Spectr 2024; 12:e0344123. [PMID: 38864649 PMCID: PMC11218442 DOI: 10.1128/spectrum.03441-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
This study aimed to characterize the composition of intestinal and nasal microbiota in septic patients and identify potential microbial biomarkers for diagnosis. A total of 157 subjects, including 89 with sepsis, were enrolled from the affiliated hospital. Nasal swabs and fecal specimens were collected from septic and non-septic patients in the intensive care unit (ICU) and Department of Respiratory and Critical Care Medicine. DNA was extracted, and the V4 region of the 16S rRNA gene was amplified and sequenced using Illumina technology. Bioinformatics analysis, statistical processing, and machine learning techniques were employed to differentiate between septic and non-septic patients. The nasal microbiota of septic patients exhibited significantly lower community richness (P = 0.002) and distinct compositions (P = 0.001) compared to non-septic patients. Corynebacterium, Staphylococcus, Acinetobacter, and Pseudomonas were identified as enriched genera in the nasal microbiota of septic patients. The constructed machine learning model achieved an area under the curve (AUC) of 89.08, indicating its efficacy in differentiating septic and non-septic patients. Importantly, model validation demonstrated the effectiveness of the nasal microecological diagnosis prediction model with an AUC of 84.79, while the gut microecological diagnosis prediction model had poor predictive performance (AUC = 49.24). The nasal microbiota of ICU patients effectively distinguishes sepsis from non-septic cases and outperforms the gut microbiota. These findings have implications for the development of diagnostic strategies and advancements in critical care medicine.IMPORTANCEThe important clinical significance of this study is that it compared the intestinal and nasal microbiota of sepsis with non-sepsis patients and determined that the nasal microbiota is more effective than the intestinal microbiota in distinguishing patients with sepsis from those without sepsis, based on the difference in the lines of nasal specimens collected.
Collapse
Affiliation(s)
- XiLan Tan
- Division of Infection Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic Testing, The department of laboratory medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zewen Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Ge
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuemei Luo
- State Key Laboratory of Organ Failure Research, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Nianyi Zeng
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Manjun Chen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiqi Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shumin Cai
- Department of Intensive Care Medicine, Nanfang Hospital, Southern Medical University, Guagnzhou, China
| | - Jun Long
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongran Cen
- Division of Intensive Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Su
- Chronic Airways Diseases Laboratory, Department of Respiratory & Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaolong He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Estrada R, Romero Y, Figueroa D, Coila P, Hañari-Quispe RD, Aliaga M, Galindo W, Alvarado W, Casanova D, Quilcate C. Effects of Age in Fecal Microbiota and Correlations with Blood Parameters in Genetic Nucleus of Cattle. Microorganisms 2024; 12:1331. [PMID: 39065099 PMCID: PMC11279168 DOI: 10.3390/microorganisms12071331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
This study aimed to determine the impact of age on the fecal microbiota in the genetic nucleus of cattle, with a focus on microbial richness, composition, functional diversity, and correlations with blood parameters. Fecal and blood samples from 21 cattle were analyzed using 16S rRNA gene sequencing. Older cattle exhibited greater bacterial diversity and abundance, with significant changes in alpha diversity indices (p < 0.05). Beta diversity analysis revealed significant variations in microbial composition between age groups and the interaction of age and sex (p < 0.05). Correlations between alpha diversity, community composition, and hematological values highlighted the influence of microbiota on bovine health. Beneficial butyrate-producing bacteria, such as Ruminococcaceae, were more abundant in older cattle, suggesting a role in gut health. Functional diversity analysis indicated that younger cattle had significantly more abundant metabolic pathways in fermentation and anaerobic chemoheterotrophy. These findings suggest management strategies including tailored probiotic therapies, dietary adjustments, and targeted health monitoring to enhance livestock health and performance. Further research should include comprehensive metabolic analyses to better correlate microbiota changes with age-related variations, enhancing understanding of the complex interactions between microbiota, age, and reproductive status.
Collapse
Affiliation(s)
- Richard Estrada
- Dirección de Desarrollo Tecnológico Agrario, Instituto Nacional de Innovación Agraria (INIA), Lima 15024, Peru; (Y.R.); (D.F.); (D.C.)
| | - Yolanda Romero
- Dirección de Desarrollo Tecnológico Agrario, Instituto Nacional de Innovación Agraria (INIA), Lima 15024, Peru; (Y.R.); (D.F.); (D.C.)
| | - Deyanira Figueroa
- Dirección de Desarrollo Tecnológico Agrario, Instituto Nacional de Innovación Agraria (INIA), Lima 15024, Peru; (Y.R.); (D.F.); (D.C.)
| | - Pedro Coila
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional del Altiplano, Puno 21001, Peru; (P.C.); (R.D.H.-Q.); (M.A.); (W.G.)
| | - Renán Dilton Hañari-Quispe
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional del Altiplano, Puno 21001, Peru; (P.C.); (R.D.H.-Q.); (M.A.); (W.G.)
| | - Mery Aliaga
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional del Altiplano, Puno 21001, Peru; (P.C.); (R.D.H.-Q.); (M.A.); (W.G.)
| | - Walter Galindo
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional del Altiplano, Puno 21001, Peru; (P.C.); (R.D.H.-Q.); (M.A.); (W.G.)
| | - Wigoberto Alvarado
- Facultad de Ingeniería Zootecnista, Agronegocios y Biotecnología, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas (UNTRM), Chachapoyas 01001, Peru;
| | - David Casanova
- Dirección de Desarrollo Tecnológico Agrario, Instituto Nacional de Innovación Agraria (INIA), Lima 15024, Peru; (Y.R.); (D.F.); (D.C.)
| | - Carlos Quilcate
- Dirección de Desarrollo Tecnológico Agrario, Instituto Nacional de Innovación Agraria (INIA), Lima 15024, Peru; (Y.R.); (D.F.); (D.C.)
| |
Collapse
|
15
|
Shi Z, Lan Y, Wang Y, Yan X, Ma X, Hassan FU, Rushdi HE, Xu Z, Wang W, Deng T. Multi-omics strategy reveals potential role of antimicrobial resistance and virulence factor genes responsible for Simmental diarrheic calves caused by Escherichia coli. mSystems 2024; 9:e0134823. [PMID: 38742910 PMCID: PMC11237395 DOI: 10.1128/msystems.01348-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
Escherichia coli (E. coli) is reported to be an important pathogen associated with calf diarrhea. Antibiotic resistance genes (ARGs) and virulence factor genes (VFGs) pose a considerable threat to both animal and human health. However, little is known about the characterization of ARGs and VFGs presented in the gut microbiota of diarrheic calves caused by E. coli. In this study, we used multi-omics strategy to analyze the ARG and VFG profiles of Simmental calves with diarrhea caused by E. coli K99. We found that gut bacterial composition and their microbiome metabolic functions varied greatly in diarrheic calves compared to healthy calves. In total, 175 ARGs were identified, and diarrheal calves showed a significantly higher diversity and abundance of ARGs than healthy calves. Simmental calves with diarrhea showed higher association of VFGs with pili function, curli assembly, and ferrienterobactin transport of E. coli. Co-occurrence patterns based on Pearson correlation analysis revealed that E. coli had a highly significant (P < 0.0001) correlation coefficient (>0.8) with 16 ARGs and 7 VFGs. Metabolomics analysis showed that differentially expressed metabolites in Simmental calves with diarrhea displayed a high correlation with the aforementioned ARGs and VFGs. Phylotype analysis of E. coli genomes showed that the predominant phylogroup B1 in diarrheic Simmental calves was associated with 10 ARGs and 3 VFGs. These findings provide an overview of the diversity and abundance of the gut microbiota in diarrheic calves caused by E. coli and pave the way for further studies on the mechanisms of antibiotic resistance and virulence in the calves affected with diarrhea.IMPORTANCESimmental is a well-recognized beef cattle breed worldwide. They also suffer significant economic losses due to diarrhea. In this study, fecal metagenomic analysis was applied to characterize the antibiotic resistance gene (ARG) and virulence factor gene (VFG) profiles of diarrheic Simmental calves. We identified key ARGs and VFGs correlated with Escherichia coli isolated from Simmental calves. Additionally, metabolomics analysis showed that differentially expressed metabolites in Simmental calves with diarrhea displayed a high correlation with the aforementioned ARGs and VFGs. Our findings provide an insight into the diversity and abundance of the gut microbiota in diarrheic calves caused by Escherichia coli and pave the way for further studies on the mechanisms of antibiotic resistance and virulence in the diarrheal calves from cattle hosts.
Collapse
Affiliation(s)
- Zhihai Shi
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Yali Lan
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Yazhou Wang
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Xiangzhou Yan
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Xiaoya Ma
- Guangxi Provincial Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China
| | - Faiz-Ul Hassan
- Institute of Animal and Dairy Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Hossam E Rushdi
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Zhaoxue Xu
- Institute of Animal Husbandry and Veterinary Medicine, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Wenjia Wang
- College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China
| | - Tingxian Deng
- Guangxi Provincial Key Laboratory of Buffalo Genetics, Breeding and Reproduction Technology, Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, China
| |
Collapse
|
16
|
Panzer JJ, Maples C, Meyer MP, Tillotson G, Theis KR, Chopra T. Gut microbiome alpha diversity decreases in relation to body weight, antibiotic exposure, and infection with multidrug-resistant organisms. Am J Infect Control 2024; 52:707-711. [PMID: 38176539 DOI: 10.1016/j.ajic.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND The human gastrointestinal tract is home to a dense and diverse microbiome, predominated by bacteria. Despite the conservation of critical functionality across most individuals, the composition of the gut microbiome is highly individualized, leading to differential responses to perturbations such as oral antibiotics or multidrug-resistant organism (MDRO) infection. Herein, subject responses to these perturbations based on their body weight were evaluated. METHODS Fecal samples were collected from 45 subjects at the Detroit Medical Center to evaluate the effects of perturbations on subjects' gut microbiome composition. Bacterial profiling was completed using 16S rRNA gene sequencing. RESULTS Subjects with multiple MDROs, subjects weighing greater than 80 kg infected with MDRO E coli, and subjects weighing less than 80 kg with exposure to vancomycin and carbapenem antibiotics during hospitalization had significantly decreased gut microbiome richness. CONCLUSIONS Both administration of oral antibiotics and MDRO infections decreased gut microbiome alpha diversity, but the magnitude of these gut microbiome perturbations was body weight dependent.
Collapse
Affiliation(s)
- Jonathan J Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Catherine Maples
- Department of Infectious Diseases, Wayne State University, Detroit, MI
| | | | | | - Kevin R Theis
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI.
| | - Teena Chopra
- Department of Infectious Diseases, Wayne State University, Detroit, MI.
| |
Collapse
|
17
|
Srifani A, Mirnawati M, Marlida Y, Rizal Y, Nurmiati N, Lee KW. Identification of novel probiotic lactic acid bacteria from soymilk waste using the 16s rRNA gene for potential use in poultry. Vet World 2024; 17:1001-1011. [PMID: 38911076 PMCID: PMC11188893 DOI: 10.14202/vetworld.2024.1001-1011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/22/2024] [Indexed: 06/25/2024] Open
Abstract
Background and Aim In-feed antibiotics have been used as antibiotic growth promoters (AGPs) to enhance the genetic potential of poultry. However, the long-term use of AGPs is known to lead to bacterial resistance and antibiotic residues in poultry meat and eggs. To address these concerns, alternatives to AGPs are needed, one of which is probiotics, which can promote the health of livestock without having any negative effects. In vitro probiotic screening was performed to determine the ability of lactic acid bacteria (LAB) isolated from soymilk waste to be used as a probiotic for livestock. Materials and Methods Four LAB isolates (designated F4, F6, F9, and F11) isolated from soymilk waste were used in this study. In vitro testing was performed on LAB isolates to determine their resistance to temperatures of 42°C, acidic pH, bile salts, hydrophobicity to the intestine, and ability to inhibit pathogenic bacteria. A promising isolate was identified using the 16S rRNA gene. Result All LAB isolates used in this study have the potential to be used as probiotics. On the basis of the results of in vitro testing, all isolates showed resistance to temperatures of 42°C and low pH (2.5) for 3 h (79.87%-94.44%) and 6 h (76.29%-83.39%), respectively. The survival rate at a bile salt concentration of 0.3% ranged from 73.24% to 90.39%, whereas the survival rate at a bile salt concentration of 0.5% ranged from 56.28% to 81.96%. All isolates showed the ability to attach and colonize the digestive tract with a hydrophobicity of 87.58%-91.88%. Inhibitory zones of LAB against pathogens ranged from 4.80-15.15 mm against Staphylococcus aureus, 8.85-14.50 mm against Salmonella enteritidis, and 6.75-22.25 mm against Escherichia coli. Although all isolates showed good ability as probiotics, isolate F4 showed the best probiotic ability. This isolate was identified as Lactobacillus casei strain T22 (JQ412731.1) using the 16S rRNA gene. Conclusion All isolates in this study have the potential to be used as probiotics. However, isolate F4 has the best probiotic properties and is considered to be the most promising novel probiotic for poultry.
Collapse
Affiliation(s)
- Anifah Srifani
- PMDSU Program, Graduate Program of Animal Feed and Nutrition Department, Faculty of Animal Science, Universitas Andalas, Padang, West Sumatera, Indonesia
| | - Mirnawati Mirnawati
- Department of Animal Feed and Nutrition, Faculty of Animal Science, Universitas Andalas, Padang, West Sumatera, Indonesia
| | - Yetti Marlida
- Department of Animal Feed and Nutrition, Faculty of Animal Science, Universitas Andalas, Padang, West Sumatera, Indonesia
| | - Yose Rizal
- Department of Animal Feed and Nutrition, Faculty of Animal Science, Universitas Andalas, Padang, West Sumatera, Indonesia
| | - Nurmiati Nurmiati
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Andalas, Padang, West Sumatera, Indonesia
| | - Kyung-Woo Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, South Korea
| |
Collapse
|
18
|
Martinez JM, Espírito Santo A, Ramada D, Fontes F, Medeiros R. Diagnostic accuracy of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and neutrophil-lymphocyte-to-platelet ratio biomarkers in predicting bacteremia and sepsis in immunosuppressive patients with cancer: literature review. Porto Biomed J 2024; 9:254. [PMID: 38835655 PMCID: PMC11146520 DOI: 10.1097/j.pbj.0000000000000254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/23/2024] [Accepted: 05/05/2024] [Indexed: 06/06/2024] Open
Abstract
Background This literature review explores the role of neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and neutrophil-lymphocyte-to-platelet ratio (NLPR) biomarkers, as potential indicators for predicting bacteremia and sepsis in patients with cancer. Objective Tracing the evolution of interest in this area since 2001, the aim of this review was to report a comprehensive overview of current knowledge and gaps, particularly in patients undergoing immunosuppression. Summary of Findings The literature research indicates the potential of NLR, PLR, and other biomarkers in diagnosing and predicting sepsis, with some studies emphasizing their value in mortality prediction. A specific focus on bacteremia shows the effectiveness of NLR and PLR as early indicators and prognostic tools, though mostly in noncancer patient populations. While NLR and PLR are promising in general cancer patient populations, the review addresses the challenges in applying these biomarkers to patients with neutropenic and lymphopenic cancer. The NLPR could be considered a significant biomarker for inflammation and mortality risk in various medical conditions, yet its diagnostic accuracy in patients with immunosuppressed cancer is not extensively validated. Conclusion This review offers a snapshot of the current research on biomarkers in patients with immunocompromised cancer in the sepsis and bacteremia area. More focused research on their application is necessary. This gap underscores an opportunity for future studies to enhance diagnostic and prognostic capabilities in this high-risk group.
Collapse
Affiliation(s)
- Jose Manuel Martinez
- Oncology Clinical Research Unit IPO Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Ana Espírito Santo
- Oncology Clinical Research Unit IPO Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Diana Ramada
- Oncology Nursing Research Unit IPO Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
| | - Filipa Fontes
- Approach to Precursor Lesions and Early Cancer Research Unit IPO Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
- Public Health Department and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group Research Unit IPO Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC) & RISE@CI-IPOP (Health Research Network), Porto, Portugal
| |
Collapse
|
19
|
Wesseltoft JB, Danielsen CD, Andersen AM, de Jonge N, Olsen A, Rohde PD, Kristensen TN. Feeding Drosophila gut microbiomes from young and old flies modifies the microbiome. Sci Rep 2024; 14:7799. [PMID: 38565609 PMCID: PMC10987527 DOI: 10.1038/s41598-024-58500-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024] Open
Abstract
It is becoming increasingly evident that the myriad of microbes in the gut, within cells and attached to body parts (or roots of plants), play crucial roles for the host. Although this has been known for decades, recent developments in molecular biology allow for expanded insight into the abundance and function of these microbes. Here we used the vinegar fly, Drosophila melanogaster, to investigate fitness measures across the lifetime of flies fed a suspension of gut microbes harvested from young or old flies, respectively. Our hypothesis was that flies constitutively enriched with a 'Young microbiome' would live longer and be more agile at old age (i.e. have increased healthspan) compared to flies enriched with an 'Old microbiome'. Three major take home messages came out of our study: (1) the gut microbiomes of young and old flies differ markedly; (2) feeding flies with Young and Old microbiomes altered the microbiome of recipient flies and (3) the two different microbial diets did not have any effect on locomotor activity nor lifespan of the recipient flies, contradicting our working hypothesis. Combined, these results provide novel insight into the interplay between hosts and their microbiomes and clearly highlight that the phenotypic effects of gut transplants and probiotics can be complex and unpredictable.
Collapse
Affiliation(s)
| | | | | | - Nadieh de Jonge
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Anders Olsen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Palle Duun Rohde
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
20
|
Sechovcová H, Mahayri TM, Mrázek J, Jarošíková R, Husáková J, Wosková V, Fejfarová V. Gut microbiota in relationship to diabetes mellitus and its late complications with a focus on diabetic foot syndrome: A review. Folia Microbiol (Praha) 2024; 69:259-282. [PMID: 38095802 DOI: 10.1007/s12223-023-01119-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/05/2023] [Indexed: 04/11/2024]
Abstract
Diabetes mellitus is a chronic disease affecting glucose metabolism. The pathophysiological reactions underpinning the disease can lead to the development of late diabetes complications. The gut microbiota plays important roles in weight regulation and the maintenance of a healthy digestive system. Obesity, diabetes mellitus, diabetic retinopathy, diabetic nephropathy and diabetic neuropathy are all associated with a microbial imbalance in the gut. Modern technical equipment and advanced diagnostic procedures, including xmolecular methods, are commonly used to detect both quantitative and qualitative changes in the gut microbiota. This review summarises collective knowledge on the role of the gut microbiota in both types of diabetes mellitus and their late complications, with a particular focus on diabetic foot syndrome.
Collapse
Affiliation(s)
- Hana Sechovcová
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Vídeňská, 1083, 142 20, Prague, Czech Republic
- Faculty of Agrobiology, Food and Natural Resources, Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences, Prague, Czech Republic
| | - Tiziana Maria Mahayri
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Vídeňská, 1083, 142 20, Prague, Czech Republic.
- Department of Veterinary Medicine, University of Sassari, 07100, Sassari, Italy.
| | - Jakub Mrázek
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, CAS, Vídeňská, 1083, 142 20, Prague, Czech Republic
| | - Radka Jarošíková
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jitka Husáková
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Veronika Wosková
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vladimíra Fejfarová
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
21
|
Erfanmanesh A, Beikzadeh B, Khanzadeh M, Alishahi M. Immuno-protective response of Asian seabass (Lates calcarifer) to inactivated vaccines against Streptococcus iniae and Vibrio harveyi. BMC Vet Res 2024; 20:89. [PMID: 38459562 PMCID: PMC10921715 DOI: 10.1186/s12917-024-03935-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND In this study, the protective immunity and immunogenicity of the monovalent and bivalent Streptococcus iniae and Vibrio harveyi vaccine were evaluated in Asian seabass. To analyze immune responses, 1200 Asian seabass with an average weight of 132.6 ± 25.4 g were divided into eight treatments in triplicates (50 fish per tank) as follows: S. iniae immunized by injection (SI), V. harveyi immunized by injection (VI), bivalent S. iniae and V. harveyi (SVI) immunized by injection, S. iniae immunized by immersion (SIM), V. harveyi (VIM) immunized by immersion, bivalent S. iniae and V. harvei (SVIM) immunized by immersion, phosphate-buffered saline (PBS) by injection (PBSI) and control group without vaccine administration (CTRL). Blood and serum samples were taken at the end of the 30th and 60th days. Then the vaccinated groups were challenged with two bacteria (S. iniae) and (V. harveyi) separately and mortality was recorded for 14 days. RESULTS This study reveals that there is no significant difference in the hematological parameters on the 30th and 60th days of the experiment in the vaccine-immunized groups compared to the CTRL group (P > 0.05). Meanwhile, there was no significant difference in the amount of serum albumin level, respiratory burst activity, and serum bactericidal activity in the vaccine-immunized groups compared to the CTRL group on the 30th and 60th days of the experiment (P > 0.05). Total protein on the 60th day (in the VI and SVI groups), globulin on the 30th day (in the VI and SVI groups) and the 60th day (in the VI group) compared to the CTRL and PBSI groups had a significant increase (P < 0.05). Complement activity (in the VI and SVI groups) and lysozyme (in the SI and SVI groups) increased significantly compared to the control group (P < 0.05). Serum antibody titer against S. iniae had a significant increase in the SI, VI, SVI and SVIM groups compared to the CTRL and PBSI groups (P < 0.05). Serum antibody titer against V. harveyi had a significant increase in the groups immunized with the vaccine compared to the CTRL and PBSI groups (P < 0.05). A significant increase in the relative percentage survival (RPS) following challenge with S. iniae in the SVI (86.6%), SI (83.3%,) and VI (73.3%) groups were observed compared to the CTRL (43.3%) and PBSI (40%) groups (P < 0.05). Also, a significant increase in the RPS after challenge with V. harveyi in the SVI group, VI 86.6%, SVI 83.3%, VIM 80% and SVIM 76.6% were observed compared to the CTRL (46.6%) and PBSI (50%) groups (P < 0.05). CONCLUSION Overall, the results demonstrated that the bivalent vaccine of S. iniae and V. harveywas able to produce significant immunogenicity and RPS in Asian seabass.
Collapse
Affiliation(s)
- Ahmad Erfanmanesh
- Animal Biological Product Research Group, Academic Center for Education, Culture and Research (ACECR), Tehran Organization, Tehran, Iran
| | - Babak Beikzadeh
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Khanzadeh
- Animal Biological Product Research Group, Academic Center for Education, Culture and Research (ACECR), Tehran Organization, Tehran, Iran.
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran.
| | - Mojtaba Alishahi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Centre of Excellence for Warm Water Fish Health and Disease, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
22
|
Rahman MN, Barua N, Tin MC, Dharmaratne P, Wong SH, Ip M. The use of probiotics and prebiotics in decolonizing pathogenic bacteria from the gut; a systematic review and meta-analysis of clinical outcomes. Gut Microbes 2024; 16:2356279. [PMID: 38778521 PMCID: PMC11123511 DOI: 10.1080/19490976.2024.2356279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Repeated exposure to antibiotics and changes in the diet and environment shift the gut microbial diversity and composition, making the host susceptible to pathogenic infection. The emergence and ongoing spread of AMR pathogens is a challenging public health issue. Recent evidence showed that probiotics and prebiotics may play a role in decolonizing drug-resistant pathogens by enhancing the colonization resistance in the gut. This review aims to analyze available evidence from human-controlled trials to determine the effect size of probiotic interventions in decolonizing AMR pathogenic bacteria from the gut. We further studied the effects of prebiotics in human and animal studies. PubMed, Embase, Web of Science, Scopus, and CINAHL were used to collect articles. The random-effects model meta-analysis was used to pool the data. GRADE Pro and Cochrane collaboration tools were used to assess the bias and quality of evidence. Out of 1395 citations, 29 RCTs were eligible, involving 2871 subjects who underwent either probiotics or placebo treatment to decolonize AMR pathogens. The persistence of pathogenic bacteria after treatment was 22%(probiotics) and 30.8%(placebo). The pooled odds ratio was 0.59(95% CI:0.43-0.81), favoring probiotics with moderate certainty (p = 0.0001) and low heterogeneity (I2 = 49.2%, p = 0.0001). The funnel plot showed no asymmetry in the study distribution (Kendall'sTau = -1.06, p = 0.445). In subgroup, C. difficile showed the highest decolonization (82.4%) in probiotics group. Lactobacillus-based probiotics and Saccharomyces boulardii decolonize 71% and 77% of pathogens effectively. The types of probiotics (p < 0.018) and pathogens (p < 0.02) significantly moderate the outcome of decolonization, whereas the dosages and regions of the studies were insignificant (p < 0.05). Prebiotics reduced the pathogens from 30% to 80% of initial challenges. Moderate certainty of evidence suggests that probiotics and prebiotics may decolonize pathogens through modulation of gut diversity. However, more clinical outcomes are required on particular strains to confirm the decolonization of the pathogens. Protocol registration: PROSPERO (ID = CRD42021276045).
Collapse
Affiliation(s)
- Md Nannur Rahman
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, N.T., Hong Kong (SAR), China
- Department of Food Technology and Nutritional Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Nilakshi Barua
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, N.T., Hong Kong (SAR), China
| | - Martha C.F. Tin
- Faculty of Medical Sciences, University College of London, London, UK
| | - Priyanga Dharmaratne
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, N.T., Hong Kong (SAR), China
| | - Sunny H. Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, N.T., Hong Kong (SAR), China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- Centre for Gut Microbiota, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, N.T., Hong Kong (SAR), China
| |
Collapse
|
23
|
Lear L, Inamine H, Shea K, Buckling A. Diversity loss from multiple interacting disturbances is regime-dependent. Ecol Lett 2023; 26:2056-2065. [PMID: 37847646 DOI: 10.1111/ele.14325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023]
Abstract
Anthropogenic activities expose many ecosystems to multiple novel disturbances simultaneously. Despite this, how biodiversity responds to simultaneous disturbances remains unclear, with conflicting empirical results on their interactive effects. Here, we experimentally test how one disturbance (an invasive species) affects the diversity of a community over multiple levels of another disturbance regime (pulse mortality). Specifically, we invade stably coexisting bacterial communities under four different pulse frequencies, and compare their final resident diversity to uninvaded communities under the same pulse mortality regimes. Our experiment shows that the disturbances synergistically interact, such that the invader significantly reduces resident diversity at high pulse frequency, but not at low. This work therefore highlights the need to study simultaneous disturbance effects over multiple disturbance regimes as well as to carefully document unmanipulated disturbances, and may help explain the conflicting results seen in previous multiple-disturbance work.
Collapse
Affiliation(s)
- Luke Lear
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, UK
| | - Hidetoshi Inamine
- Department of Biology and Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Katriona Shea
- Department of Biology and Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Angus Buckling
- Centre for Ecology and Conservation, University of Exeter, Penryn, Cornwall, UK
| |
Collapse
|
24
|
Liu RH, Sun AQ, Liao Y, Tang ZX, Zhang SH, Shan X, Hu JT. Lactiplantibacillus plantarum Regulated Intestinal Microbial Community and Cytokines to Inhibit Salmonella typhimurium Infection. Probiotics Antimicrob Proteins 2023; 15:1355-1370. [PMID: 36074298 DOI: 10.1007/s12602-022-09987-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/25/2022]
Abstract
Lactic acid bacteria (LAB) are recognized as food-grade safe microorganisms and have many beneficial effects. LAB could maintain the host intestinal homeostasis and regulate intestinal microbial community to exert antibacterial effects. In this study, Lactiplantibacillus plantarum (L. plantarum, Lp01) strain isolated from pig intestine was orally administered to C57BL/6 mice, and mice were then infected with Salmonella typhimurium (ATCC14028). The protective effects of L. plantarum were evaluated by monitoring body weight loss, survival rates, bacterial loads in tissue, colon histopathology analysis, and cytokine secretion. 16S rRNA gene sequencing was also utilized to detect the dynamics of the blind gut microbial community in mice. We found that L. plantarum could significantly reduce the body weight loss and improve the survival rates. The survival rate in the L. P-Sty group was up to 67.5%, which was much higher than that in the STY group (25%). Counting of bacterial loads displayed that the colony-forming unit (CFU) of S. typhimurium in the spleen (p < 0.05) and the liver (p < 0.05) from L. P-Sty group both decreased, compared with STY group. Intestinal histopathology showed that it alleviated the intestinal injury caused by Salmonella, inhibited the secretion of pro-inflammatory cytokines, and promoted anti-inflammatory cytokines (p < 0. 01). In addition, L. plantarum also significantly ameliorated the intestinal gut microbiome disturbance caused by Salmonella. It displayed an obvious increase of beneficial bacteria including Lactobacillus and Bacteroidetes and reduction of pathogenic bacteria like Proteobacteria. In conclusion, L. plantarum could regulate microbial community to inhibit Salmonella typhimurium infection.
Collapse
Affiliation(s)
- Rui-Han Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - An-Qi Sun
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ye Liao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Zheng-Xu Tang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Shi-Han Zhang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Shan
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jing-Tao Hu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
25
|
Wang X, Pan L, Wang F, Long F, Yang B, Tang D. Interventional effects of oral microecological agents on perioperative indicators of colorectal cancer: a meta-analysis. Front Oncol 2023; 13:1229177. [PMID: 37681033 PMCID: PMC10482437 DOI: 10.3389/fonc.2023.1229177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Purpose To investigate the efficacy of the application of microecological agents in patients with perioperative colorectal cancer. Methods The seven electronic databases including PubMed, Cochrane Library, Excerpt Medica Database (Embase), Web of Science (WOS), Chinese Biomedical Literature Database (CBM), China National Knowledge Infrastructure (CNKI), and Wan-fang Database were systematically searched for eligible studies from 2000 to February 2023. Results A total of 38 randomized controlled clinical trials were included in this study, with a total of 1765 patients in the microecological preparation group and 1769 patients in the control group. All data were analyzed using Review Manager 5.4 and R 4.2.2 software. Meta-analysis showed that in the perioperative period of colorectal cancer, the microecological agents group reduced patients' adverse drug reactions, improved intestinal flora with Lactobacillus (SMD, 3.0858, [2.0197; 4.1520], p< 0. 0001), Bifidobacterium (SMD, 2.1551, [1.6145; 2.6956], p< 0.0001) and Escherichia coli (SMD, -1.1393, [-1.6247; -0.6538], p< 0.0001); protection of intestinal mucosal barrier function, endotoxin (SMD, -2.6850 [-4.1399; -1.2301], p=0.0003), DAO (SMD, -2.5916, [-3.4694; -1.7137], p<0.0001) and plasma D-lactate (SMD, -5.4726, [-9.8901; -1.0551], p= 0.0152), reduced inflammatory response, IL-6 (SMD, -3.1279 [-5.7706; -0.4852], p=0.0204) and CRP (SMD, -3.9698 [-7.6296; -0.3100], p=0.0335); improved the immune function of the organism, CD4+ (SMD, 1.5817 [1.0818; 2.0817], p< 0.0001), CD4+/CD8+ (SMD, 1.2938 [0.9693; 1.6183] p< 0.0001) and IgG (SMD, 1.1376 [0.2993; 1.9759] p=0.0078), improved short-term clinical efficacy, ORR (RR, 1.5105 [1.2306; 1.8541], p< 0.0001) and DCR (RR, 0.3896 [0.2620; 0.5795], p< 0.0001). Conclusion By increasing the number of beneficial flora such as Lactobacillus and Bifidobacterium and decreasing the number of harmful flora such as Escherichia coli, the micro-ecological preparation group is beneficial in improving the ecological dysregulation in colorectal cancer patients receiving different treatments in the perioperative period. The microecological preparation group was able to reduce many types of adverse drug reactions, such as infections and gastrointestinal discomfort, compared to the control group. The microecological agents also reduced inflammatory responses, decreased the increase in harmful metabolites, enhanced patients' immune function, protected intestinal mucosal barrier function, and improved short-term clinical outcomes. Systematic review registration https://inplasy.com/inplasy-2023-4-0051/, identifier INPLASY202340051.
Collapse
Affiliation(s)
- Xueyan Wang
- The First College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Lijun Pan
- Department of Medical Affairs, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Feiqing Wang
- Research Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Fengxi Long
- Development Planning Division, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Bing Yang
- Student Management Office, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Dongxin Tang
- Dean’s Office, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
26
|
Girma A. The effect of novel antimicrobial agents on the normal functioning of human intestinal microbiota: a systematic review. FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1159352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Antimicrobial agents have significant effects on the ecological balance of the human microbiota through incomplete absorption (e.g., orally administered antimicrobial agents) or secretion (e.g., by the salivary glands, in the bile, or from the intestinal mucosa) of the agents. This study aimed to examine the effects of novel antimicrobial agents on the normal functioning of the intestinal microbiota. The articles, written in English, were recovered from PubMed, ScienceDirect, Web of Science, Google Scholar, and DOAJ, as well as from manual searches using a reference list. “Microbiota”, “Intestinal Microbiota”, “Eubiotic Microbiota”, “Ecological Impact”, “Antimicrobial Agents,”, “Antibiotics”, “Dysbiosis”, “Gut Microbiota”, and “Probiotics” were the search terms used to retrieve the articles. The PRISMA 2009 checklist was applied for article search strategy, article selection, data extraction, and result reporting for the review process. A total of eight original research articles were included from a total of 379 articles obtained in different search strategies. The eight new antimicrobial agents demonstrated significant impacts on the ecological balance of the human intestinal microbiota. Therefore, eubiosis is crucial in preventing the establishment of exogenous antimicrobial-resistant strains as well as their gene transfer.Systematic review registration[PRISMA], identifier [2009].
Collapse
|
27
|
Wang M, Qin Y, Liu Y, Yang H, Wang J, Ru S, Cui P. Short-term exposure to enrofloxacin causes hepatic metabolism disorder associated with intestinal flora dysbiosis in adult marine medaka (Oryzias melastigma). MARINE POLLUTION BULLETIN 2023; 192:114966. [PMID: 37178644 DOI: 10.1016/j.marpolbul.2023.114966] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023]
Abstract
Enrofloxacin (ENR) is a widely used fluoroquinolone antibiotic that is frequently detected in the environment. Our study assessed the impact of short-term ENR exposure on the intestinal and liver health of marine medaka (Oryzias melastigma) using gut metagenomic shotgun sequencing and liver metabolomics. We found that ENR exposure resulted in imbalances of Vibrio and Flavobacteria and enrichments of multiple antibiotic resistance genes. Additionally, we found a potential link between the host's response to ENR exposure and the intestinal microbiota disorder. Liver metabolites, including phosphatidylcholine, lysophosphatidylcholine, taurocholic acid, and cholic acid, in addition to several metabolic pathways in the liver that are closely linked to the imbalance of intestinal flora were severely maladjusted. These findings suggest that ENR exposure has the potential to negatively affect the gut-liver axis as the primary toxicological mechanism. Our findings provide evidence regarding the negative physiological impacts of antibiotics on marine fish.
Collapse
Affiliation(s)
- Meiru Wang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Yifan Qin
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Yifan Liu
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Hui Yang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Jun Wang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China
| | - Pengfei Cui
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao, Shandong, China.
| |
Collapse
|
28
|
Senchukova MA. Microbiota of the gastrointestinal tract: Friend or foe? World J Gastroenterol 2023; 29:19-42. [PMID: 36683718 PMCID: PMC9850957 DOI: 10.3748/wjg.v29.i1.19] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/05/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023] Open
Abstract
The gut microbiota is currently considered an external organ of the human body that provides important mechanisms of metabolic regulation and protection. The gut microbiota encodes over 3 million genes, which is approximately 150 times more than the total number of genes present in the human genome. Changes in the qualitative and quantitative composition of the microbiome lead to disruption in the synthesis of key bacterial metabolites, changes in intestinal barrier function, and inflammation and can cause the development of a wide variety of diseases, such as diabetes, obesity, gastrointestinal disorders, cardiovascular issues, neurological disorders and oncological concerns. In this review, I consider issues related to the role of the microbiome in the regulation of intestinal barrier function, its influence on physiological and pathological processes occurring in the body, and potential new therapeutic strategies aimed at restoring the gut microbiome. Herewith, it is important to understand that the gut microbiota and human body should be considered as a single biological system, where change of one element will inevitably affect its other components. Thus, the study of the impact of the intestinal microbiota on health should be considered only taking into account numerous factors, the role of which has not yet been fully elucidated.
Collapse
Affiliation(s)
- Marina A Senchukova
- Department of Oncology, Orenburg State Medical University, Orenburg 460000, Russia
| |
Collapse
|
29
|
Dahdouh E, Cendejas-Bueno E, Ruiz-Carrascoso G, Schüffelmann C, Lázaro-Perona F, Castro-Martínez M, Moreno-Ramos F, Escosa-García L, Alguacil-Guillén M, Mingorance J. Intestinal loads of extended-spectrum beta-lactamase and Carbapenemase genes in critically ill pediatric patients. Front Cell Infect Microbiol 2023; 13:1180714. [PMID: 37201116 PMCID: PMC10188119 DOI: 10.3389/fcimb.2023.1180714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/13/2023] [Indexed: 05/20/2023] Open
Abstract
Introduction Intestinal colonization by Multi-Drug Resistant Organisms (MDROs) can pose a threat on the health of critically ill patients. The extent of colonization by these organisms is related to previous antibiotic treatments and their ability to cause infections among adult patients. The aim of this study is to determine the relationship between the intestinal Relative Loads (RLs) of selected antibiotic resistance genes, antibiotic consumption and extra-intestinal spread among critically ill pediatric patients. Methods RLs of bla CTX-M-1-Family, bla OXA-1, bla OXA-48 and bla VIM were determined in 382 rectal swabs obtained from 90 pediatric critically ill patients using qPCRs. The RLs were compared to the patients' demographics, antibiotic consumption, and detection of MDROs from extra-intestinal sites. 16SrDNA metagenomic sequencing was performed for 40 samples and clonality analyses were done for representative isolates. Results and discussion 76 (74.45%) patients from which 340 (89.01%) rectal swabs were collected had at least one swab that was positive for one of the tested genes. Routine cultures did not identify carbapenemases in 32 (45.1%) and 78 (58.2%) swabs that were positive by PCR for bla OXA-48 and blaVIM, respectively. RLs of above 6.5% were associated with extra-intestinal spread of blaOXA-48-harboring MDROs. Consumption of carbapenems, non-carbapenem β-lactams, and glycopeptides were statistically associated with testing negative for bla CTX-M-1-Family and bla OXA-1 while the consumption of trimethoprim/sulfamethoxazole and aminoglycosides was associated with testing negative for blaOXA-48 (P<0.05). In conclusion, targeted qPCRs can be used to determine the extent of intestinal dominance by antibiotic resistant opportunistic pathogens and their potential to cause extra-intestinal infections among a critically ill pediatric population.
Collapse
Affiliation(s)
- Elias Dahdouh
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- *Correspondence: Elias Dahdouh,
| | - Emilio Cendejas-Bueno
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo Ruiz-Carrascoso
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Fernando Lázaro-Perona
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | | | | | - Luis Escosa-García
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Tropical and Infectious Diseases Department, Hospital Universitario La Paz, Madrid, Spain
| | - Marina Alguacil-Guillén
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Jesús Mingorance
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Delivery of streptomycin to the rat colon by use of electrospun nanofibers. Sci Rep 2022; 12:21503. [PMID: 36513721 PMCID: PMC9747919 DOI: 10.1038/s41598-022-25769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Drug-loaded electrospun nanofibers are potential drug carrier systems that may optimize disease treatment while reducing the impact on commensal microbes. The feasibility of streptomycin-loaded pullulan nanofibers fabricated from a green electrospinning procedure using water as the solvent was assessed. We conducted a rat study including a group treated with streptomycin-loaded nanofibers (STR-F, n = 5), a group treated with similar concentrations of streptomycin in the drinking water (STR-W, n = 5), and a non-treated control group (CTR, n = 5). Streptomycin was successfully loaded into nanofibers and delivered by this vehicle, which minimized the quantity of the drug released in the ileal compartment of the gut. Ingested streptomycin-resistant E. coli colonized of up to 106 CFU/g feces, revealing a selective effect of streptomycin even when given in the low amounts allowed by the nanofiber-based delivery. 16S amplicon sequencing of the indigenous microbiota revealed differential effects in the three groups. An increase of Peptostreptococcaceae in the cecum of STR-F animals may indicate that the fermentation of nanofibers directly or indirectly promoted growth of bacteria within this family. Our results elucidate relevant properties of electrospun nanofibers as a novel vehicle for delivery of antimicrobials to the large intestine.
Collapse
|
31
|
Huertas-Abril PV, Prieto-Álamo MJ, Jurado J, García-Barrera T, Abril N. A selenium-enriched diet helps to recover liver function after antibiotic administration in mice. Food Chem Toxicol 2022; 171:113519. [PMID: 36464106 DOI: 10.1016/j.fct.2022.113519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/20/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
Antibiotic (Abx) treatments or inadvertent exposure to Abx-contaminated food and water can adversely affect health. Many studies show strong correlations between Abx and liver damage pointing to gut dysbiosis as a contributing factor because the gut microbiota (GM) forms a complex network with liver. Selenium (Se) is a beneficial micronutrient able to shape the composition of the GM. We analyzed here the ability of a low dose (120 μg/kg bodyweight/day) Se-enriched diet to ameliorate the effects of a 7-day intervention with an Abx-cocktail over the global health and the homeostasis of cholesterol and bile acids in the mouse liver. We found that Se restored lipid metabolism preventing the increased synthesis and accumulation of cholesterol caused by Abx treatment. Integrating these results with previous metataxonomic and metabolomic data in same mice, we conclude that part of the effect of Se against liver dysfunction (cholesterol and bile acids metabolism and transport) could be mediated by the GM. We provide data that contribute to a more complete view of the molecular mechanisms underlying the beneficial action of Se on health, pointing to a possible use of low doses of Se as a functional food additive (prebiotic) to prevent the negative effects of antibiotics.
Collapse
Affiliation(s)
- Paula V Huertas-Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - María-José Prieto-Álamo
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| | - Juan Jurado
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Tamara García-Barrera
- Research Center of Natural Resources, Health and the Environment (RENSMA). Department of Chemistry, Faculty of Experimental Sciences, Campus El Carmen, University of Huelva, Fuerzas Armadas Ave., 21007, Huelva, Spain
| | - Nieves Abril
- Department of Biochemistry and Molecular Biology, University of Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|
32
|
Olbertz D, Proquitté H, Patzer L, Erler T, Mikolajczak A, Sadowska-Krawczenko I, Wolff C, Radke M. Potential Benefit of Probiotic E. Coli Nissle in Term Neonates. KLINISCHE PADIATRIE 2022. [PMID: 36446590 PMCID: PMC10328724 DOI: 10.1055/a-1970-4340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Probiotics are often viewed as an immunity enhancing agent. The objective of this study was to investigate whether oral administration of Escherichia coli Nissle 1917 reduces the number of infections, their duration, and severity in the first 24 months after parturition in healthy neonates. SUBJECTS AND METHODS This prospective, confirmatory, randomised, double-blind, placebo-controlled study enrolled 567 healthy neonates from four German and two Polish sites. Neonates received 10e8 viable E. coli Nissle (n=283) or placebo (n=284) daily in the first week and every second day in week 2 and 3. After 6 and 12 months, the subjects received additional instillations on ten subsequent days. The overall efficacy was assessed by the number of infections per observation period. RESULTS Incidence rates of infection, infection duration and severity showed no statistically significant difference between groups after 24 months. Post-hoc analyses, however, revealed a short-term benefit of E. coli Nissle four weeks after treatment start which became less pronounced after eight weeks. E. coli Nissle was safe and well tolerated. CONCLUSIONS A long-term effect after colonising the healthy neonate´s gut with E. coli Nissle to protect against infections could not be shown. Additional studies are needed to confirm a transitory, yet clinically significant role of probiotics in the first four weeks after parturition.
Collapse
Affiliation(s)
- Dirk Olbertz
- Department of Neonatology, Klinikum Südstadt Rostock, Rostock, Germany
| | - Hans Proquitté
- Section of Neonatology and Neonatal Intensive Care, Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Ludwig Patzer
- Pediatrics, Children's Hospital St. Elisabeth and St. Barbara, Halle/Saale, Germany
| | - Thomas Erler
- Department of Pediatrics, Klinikum Westbrandenburg, Potsdam, Germany
| | - Alexsandra Mikolajczak
- Department of Neonatology, University Clinical Centre of Warsaw Medical University, Warsaw, Poland
| | - Iwona Sadowska-Krawczenko
- Department of Neonatology, Nicolaus Copernicus University in Toruń Ludwik Rydygier Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Corinna Wolff
- Department of Clinical Research, Ardeypharm GmbH, Herdecke, Germany
| | - Michael Radke
- Department of Pediatrics, University Hospital Rostock, Rostock, Germany
| |
Collapse
|
33
|
Singh S, Singh M, Gaur S. Probiotics as multifaceted oral vaccines against colon cancer: A review. Front Immunol 2022; 13:1002674. [PMID: 36263037 PMCID: PMC9573965 DOI: 10.3389/fimmu.2022.1002674] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics are known as the live microorganisms that, upon adequate administration, elicit a health beneficial response inside the host. The probiotics are known as immunomodulators and exhibit anti-tumor properties. Advanced research has explored the potential use of probiotics as the oral vaccines without the latent risks of pathogenicity. Probiotic-based oral vaccines are known to induce mucosal immunity that prevents the host from several enteric infections. Probiotic bacteria have the ability to produce metabolites in the form of anti-inflammatory cytokines, which play an important role in the prevention of carcinogenesis and in the activation of the phagocytes that eliminate the preliminary stage cancer cells. This review discusses the advantages and disadvantages of using the oral probiotic vaccines as well as the mechanism of action of probiotics in colon cancer therapy. This review also employs the use of “PROBIO” database for selecting certain probiotics with immunomodulatory properties. Furthermore, the use of several probiotic bacteria as anti-colon cancer adjuvants has also been discussed in detail. Because the current studies and trials are more focused on using the attenuated pathogens instead of using the probiotic-based vaccines, future studies must involve the advanced research in exploiting the potential of several probiotic strains as adjuvants in cancer therapies.
Collapse
Affiliation(s)
- Shubhi Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Manisha Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- *Correspondence: Smriti Gaur,
| |
Collapse
|
34
|
Li X, Jing K, Lu H, Li K, Zhang Y, Hasichaolu. Exploring the Correlation between Changes in Gut Microbial Community Diversity and Depression in Human Populations. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6334868. [PMID: 35937392 PMCID: PMC9355758 DOI: 10.1155/2022/6334868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/12/2022] [Indexed: 11/18/2022]
Abstract
Depression, also known as depressive disorder, is a group of psychosomatic affective disorders characterized by persistent and significantly depressed mood, delayed thinking, and cognitive impairment. The aim of this study was to explore the correlation between changes in gut microbial community diversity and depression to provide data on new strategies for the prevention and treatment of depression. In this study, we separated participants into a group of depressed patients and a healthy comparison group. We analyzed the gut microbial community structure of depressed patients and healthy comparisons using second-generation sequencing of the bacterial 16S RNA gene. There were significant differences in the gut microflora structure between patients with depression and healthy individuals. The gut flora alpha diversity index was significantly reduced in patients with depression compared to that in the healthy population. At the species level, the relative abundance of Coprococcus catus and Bacteroides barnesiae was significantly lower in the depressed group than that in the control group. The development of depression may be associated with a decrease in beneficial gut bacteria.
Collapse
Affiliation(s)
- Xin Li
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ke Jing
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hong Lu
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ke Li
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yaowu Zhang
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Hasichaolu
- Department of Laboratory Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
35
|
Gough EK. The impact of mass drug administration of antibiotics on the gut microbiota of target populations. Infect Dis Poverty 2022; 11:76. [PMID: 35773678 PMCID: PMC9245274 DOI: 10.1186/s40249-022-00999-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Antibiotics have become a mainstay of healthcare in the past century due to their activity against pathogens. This manuscript reviews the impact of antibiotic use on the intestinal microbiota in the context of mass drug administration (MDA). The importance of the gut microbiota to human metabolism and physiology is now well established, and antibiotic exposure may impact host health via collateral effects on the microbiota and its functions. To gain further insight into how gut microbiota respond to antibiotic perturbation and the implications for public health, factors that influence the impact of antibiotic exposure on the microbiota, potential health outcomes of antibiotic-induced microbiota alterations, and strategies that have the potential to ameliorate these wider antibiotic-associated microbiota perturbations are also reviewed.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of International Health, Human Nutrition Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
36
|
Wang Z, Liu J, Li F, Luo Y, Ge P, Zhang Y, Wen H, Yang Q, Ma S, Chen H. The gut-lung axis in severe acute Pancreatitis-associated lung injury: The protection by the gut microbiota through short-chain fatty acids. Pharmacol Res 2022; 182:106321. [PMID: 35752356 DOI: 10.1016/j.phrs.2022.106321] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/15/2022] [Accepted: 06/18/2022] [Indexed: 02/07/2023]
Abstract
The role of gut microbiota in regulating the intestinal homeostasis, as well as the pathogenesis of severe acute pancreatitis-associated lung injury (PALI) is widely recognized. The bioactive functions of metabolites with small molecule weight and the detail molecular mechanisms of PALI mediated by "gut-lung axis" have gradually raised the attentions of researchers. Several studies have proved that short-chain fatty acids (SCFAs) produced by gut microbiome play crucial roles and varied activities in the process of PALI. However, relevant reviews reporting SCFAs in the involvement of PALI is lacking. In this review, we firstly introduced the synthetic and metabolic pathways of SCFAs, as well as the transport and signal transduction routes in brief. Afterwards, we focused on the possible mechanisms and clues of SCFAs to participate in the fight against PALI which referred to the inhibition of pathogen proliferation, anti-inflammatory effects, enhancement of intestinal barrier functions, and the maintenance and regulation of immune homeostasis via pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). In addition, the latest reported pathological and physiological mechanisms of the gut-lung axis involved in PALI were reviewed. Finally, we summarized the potential therapeutic interventions of PALI by targeting SCFAs, including dietary fiber supplementation, direct supplementation of SCFAs/prebiotics/probiotics, and drugs administration, which is expected to provide new sights for clinical use in the future.
Collapse
Affiliation(s)
- Zhengjian Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Fan Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Peng Ge
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Yibo Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Haiyun Wen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Qi Yang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China.
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China; Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, PR China.
| |
Collapse
|
37
|
Effect of Probiotic-Assisted Eradication of cagA+/vacA s1m1 Helicobacter pylori on Intestinal Flora. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8607671. [PMID: 35528160 PMCID: PMC9076325 DOI: 10.1155/2022/8607671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 12/14/2022]
Abstract
Objective. We attempted to evaluate the effects of probiotic-assisted eradication of cytotoxin-associated gene A (cagA)+/vacuolating cytotoxin A (vacA) s1m1 Helicobacter pylori (H. pylori) on the intestinal flora, inflammatory factors, and clinical outcomes. Methods. A total of 180 patients with cagA+/vacA s1m1 H. pylori were randomly divided into two groups. Group A was treated with bismuth quadruple therapy (BQT). Group B was treated with S. boulardii in addition to BQT. The distribution of intestinal flora, serum interleukin-8 (IL-8), IL-17, tumor necrosis factor-α (TNF-α) levels, recovery time of clinical symptoms, total effective rate of clinical symptoms, H. pylori eradication rate, and adverse reactions were observed. Results. 2 weeks after treatment, the contents of Bifidobacterium, Bacteroides, and Lactobacillus in the intestinal tract of Group A decreased, while the amounts of Enterococcus and Enterobacter increased. In Group B, the contents of Bifidobacterium, Bacteroides, and Lactobacillus increased, while the amounts of Enterococcus and Enterobacter did not change significantly. Moreover, the trend of this flora change was still present at 4 weeks after treatment. Compared with Group A, Group B had lower IL-8, IL-17, and TNF-α levels, shorter recovery time of clinical symptoms, higher overall efficiency of clinical symptoms, and lower occurrence of adverse reactions. The eradication rate did not differ significantly between the two groups. Conclusion. BQT can lead to intestinal flora disorders in cagA+/vacA s1m1 H. pylori patients. S. boulardii can improve the distribution of intestinal flora, downregulate immune-inflammatory mediators, and modify clinical symptoms in patients.
Collapse
|
38
|
The Nutrition-Microbiota-Physical Activity Triad: An Inspiring New Concept for Health and Sports Performance. Nutrients 2022; 14:nu14050924. [PMID: 35267899 PMCID: PMC8912693 DOI: 10.3390/nu14050924] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
The human gut microbiota is currently the focus of converging interest in many diseases and sports performance. This review presents gut microbiota as a real “orchestra conductor” in the host’s physio(patho)logy due to its implications in many aspects of health and disease. Reciprocally, gut microbiota composition and activity are influenced by many different factors, such as diet and physical activity. Literature data have shown that macro- and micro-nutrients influence gut microbiota composition. Cumulative data indicate that gut bacteria are sensitive to modulation by physical activity, as shown by studies using training and hypoactivity models. Sports performance studies have also presented interesting and promising results. Therefore, gut microbiota could be considered a “pivotal” organ for health and sports performance, leading to a new concept: the nutrition-microbiota-physical activity triad. The next challenge for the scientific and medical communities is to test this concept in clinical studies. The long-term aim is to find the best combination of the three elements of this triad to optimize treatments, delay disease onset, or enhance sports performance. The many possibilities offered by biotic supplementation and training modalities open different avenues for future research.
Collapse
|
39
|
Di Marcantonio L, Marotta F, Vulpiani MP, Sonntag Q, Iannetti L, Janowicz A, Serafino GD, Di Giannatale E, Garofolo G. Investigating the cecal microbiota in broiler poultry farms and its potential relationships with animal welfare. Res Vet Sci 2022; 144:115-125. [DOI: 10.1016/j.rvsc.2022.01.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
|
40
|
Gut microbiome in the emergence of antibiotic-resistant bacterial pathogens. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:1-31. [DOI: 10.1016/bs.pmbts.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
41
|
Serek P, Oleksy-Wawrzyniak M. The Effect of Bacterial Infections, Probiotics and Zonulin on Intestinal Barrier Integrity. Int J Mol Sci 2021; 22:11359. [PMID: 34768787 PMCID: PMC8583036 DOI: 10.3390/ijms222111359] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal barrier plays an extremely important role in maintaining the immune homeostasis of the gut and the entire body. It is made up of an intricate system of cells, mucus and intestinal microbiota. A complex system of proteins allows the selective permeability of elements that are safe and necessary for the proper nutrition of the body. Disturbances in the tightness of this barrier result in the penetration of toxins and other harmful antigens into the system. Such events lead to various digestive tract dysfunctions, systemic infections, food intolerances and autoimmune diseases. Pathogenic and probiotic bacteria, and the compounds they secrete, undoubtedly affect the properties of the intestinal barrier. The discovery of zonulin, a protein with tight junction regulatory activity in the epithelia, sheds new light on the understanding of the role of the gut barrier in promoting health, as well as the formation of diseases. Coincidentally, there is an increasing number of reports on treatment methods that target gut microbiota, which suggests that the prevention of gut-barrier defects may be a viable approach for improving the condition of COVID-19 patients. Various bacteria-intestinal barrier interactions are the subject of this review, aiming to show the current state of knowledge on this topic and its potential therapeutic applications.
Collapse
Affiliation(s)
- Paweł Serek
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Oleksy-Wawrzyniak
- Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| |
Collapse
|
42
|
The role of microbiota in respiratory health and diseases, particularly in tuberculosis. Biomed Pharmacother 2021; 143:112108. [PMID: 34560539 DOI: 10.1016/j.biopha.2021.112108] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022] Open
Abstract
Trillions of beneficial and hostile microorganisms live in the human respiratory and gastrointestinal tracts, which act as gatekeepers in maintaining human health, i.e., protecting the body from pathogens by colonizing mucosal surfaces with microbiota-derived antimicrobial metabolites such as short-chain fatty acids or host-derived cytokines and chemokines. It is widely accepted that the microbiome interacts with each other and with the host in a mutually beneficial relationship. Microbiota in the respiratory tract may also play a crucial role in immune homeostasis, maturation, and maintenance of respiratory physiology. Anti-TB antibiotics may cause dysbiosis in the lung and intestinal microbiota, affecting colonization resistance and making the host more susceptible to Mycobacterium tuberculosis (M. tuberculosis) infection. This review discusses recent advances in our understanding of the lung microbiota composition, the lungs and intestinal microbiota related to respiratory health and diseases, microbiome sequencing and analysis, the bloodstream, and the lymphatic system that underpin the gut-lung axis in M. tuberculosis-infected humans and animals. We also discuss the gut-lung axis interactions with the immune system, the role of the microbiome in TB pathogenesis, and the impact of anti-TB antibiotic therapy on the microbiota in animals, humans, and drug-resistant TB individuals.
Collapse
|