1
|
Rezaei AR, Ates F, Sulik A, Toczyłowski K. 'Smart', microbiome-sparing antibacterial therapy with a focus on the novel Lolamicin: an overview. Infection 2025:10.1007/s15010-025-02538-4. [PMID: 40220252 DOI: 10.1007/s15010-025-02538-4] [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: 01/07/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
PURPOSE Antibiotic resistance (AR) is an escalating worldwide health emergency, requiring inventive strategies for antibiotic treatment. This review examines the tactics used in designing smart antibiotics, with a specific emphasis on the mechanism of action of lolamicin, a newly developed microbiome-sparing antibiotic. METHODS We review the recent advances in smart antibiotic development, particularly those aiming to preserve the gut microbiome while effectively targeting pathogens. The study focuses on lolamicin's selective targeting mechanism, its inhibition of the LolCDE complex in Gram-negative bacteria. RESULTS Lolamicin works by blocking the LolCDE complex, which is crucial for transporting lipoproteins in Gramnegative bacteria. It offers a significant improvement compared to conventional antibiotics and other microbiomesparing options by safeguarding the microbiome and reducing the development of resistance. However, its limited range of effectiveness - namely against certain harmful bacteria such as Pseudomonas aeruginosa - and the possibility of bacteria becoming resistant to it, remain areas of concern. CONCLUSION Lolamicin presents a hopeful resolution by selectively attacking Gram-negative bacteria while leaving the beneficial gut flora unharmed. Further investigation and rigorous clinical testing are essential to fully harness its promise and confirm its long-term utility in combating antibiotic resistance.
Collapse
Affiliation(s)
- Ahmad Reza Rezaei
- 1Department of Pediatric Infectious Diseases, Medical University of Bialystok, Białystok, Poland
| | - Furkan Ates
- 1Department of Pediatric Infectious Diseases, Medical University of Bialystok, Białystok, Poland
| | - Artur Sulik
- 1Department of Pediatric Infectious Diseases, Medical University of Bialystok, Białystok, Poland
| | - Kacper Toczyłowski
- 1Department of Pediatric Infectious Diseases, Medical University of Bialystok, Białystok, Poland.
| |
Collapse
|
2
|
Lu J, Wu H, Wu S, Wang S, Fan H, Ruan H, Qiao J, Caiyin Q, Wen M. Salmonella: Infection mechanism and control strategies. Microbiol Res 2025; 292:128013. [PMID: 39675139 DOI: 10.1016/j.micres.2024.128013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Salmonella is a foodborne pathogen that predominantly resides in the intestinal tract of humans and animals. Infections caused by Salmonella can lead to various illnesses, including gastroenteritis, bacteremia, septicemia, and focal infections, with severe cases potentially resulting in host mortality. The mechanisms by which Salmonella invades host cells and disseminates throughout the body are partly understood, but there are still many scientific questions to be solved. This review aims to synthesize existing research on the interactions between Salmonella and hosts, detailing a comprehensive infection mechanism from adhesion and invasion to intracellular propagation and systemic spread. Overuse of antibiotics contributes to the emergence of drug-resistant Salmonella strains. An in-depth analysis of the mechanism of Salmonella infection will provide a theoretical basis for the development of novel Salmonella control strategies. These innovative control strategies include antibiotic adjuvants, small molecules, phages, attenuated vaccines, and probiotic therapies, which show huge potential in controlling Salmonella infection.
Collapse
Affiliation(s)
- Juane Lu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Wu
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Shengli Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Hongfei Fan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Haihua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Mingzhang Wen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
3
|
Ghosh PR, Al Hasan MS, Rouf R, Chowdhury R, Yadav B, Mia E, Islam MT, Hasan MR, Ansari SA, Ansari IA, Bhuia MS, Islam MT. Assessments of protodioscin's antinociceptive and antidiarrheal properties: in vivo and in silico investigations on macromolecule binding affinity and modulatory effects. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03860-2. [PMID: 39921733 DOI: 10.1007/s00210-025-03860-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Protodioscin (PRO) is a furostanol saponin with antioxidant and anti-inflammatory properties. However, there is no proof against nociception and diarrhea. The study aims to investigate the antinociceptive and antidiarrheal effects of PRO, comparing its efficacy with diclofenac sodium (DFS) and loperamide (LOP) using in vivo and in silico methods. Antinociceptive activity was evaluated using the acetic acid-induced writhing and formalin-induced paw licking tests, and antidiarrheal effects were assessed via castor oil-induced diarrhea in mice. Mice were divided into groups receiving PRO (2.5 and 10 mg/kg, p.o.), DFS (25 mg/kg, p.o.), LOP (3 mg/kg, p.o.), or combinations. Molecular docking studies were conducted on COX-1, COX-2 enzymes, and the Mu-opioid receptor (MOR), with toxicity predictions performed for safety profiling. In vivo results demonstrated that PRO significantly (p < 0.05) reduced pain and diarrhea in animals. PRO at 10 mg/kg, showed comparable efficacy to DFS and LOP (25 and 3 mg/kg) in both models. Molecular docking revealed that PRO had stronger binding affinities with COX-1 (‒10.0 kcal/mol), COX-2 (‒9.6 kcal/mol) enzymes, and MOR (‒7.7 kcal/mol) compared to standard drugs. Toxicity predictions indicate PRO is relatively safe in some toxicity parameters. PRO exhibits significant antinociceptive and antidiarrheal activities comparable to DFS and LOP, making it a promising natural alternative for managing pain and diarrhea. Additional clinical trials and pharmacokinetic assessments are required to evaluate its long-term safety for use.
Collapse
Affiliation(s)
- Pompa Rani Ghosh
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Bangladesh
| | - Razina Rouf
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Bangladesh
| | - Balkrishnan Yadav
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Emon Mia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Bangladesh
| | - Md Tanvir Islam
- Department of Chemistry, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Rakibul Hasan
- Department of Chemistry, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10124, Turin, Italy
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Bangladesh
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, 8100, Bangladesh.
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh.
| |
Collapse
|
4
|
Marcari AB, Paiva AD, Simon CR, Dos Santos MESM. Leaky Gut Syndrome: An Interplay Between Nutrients and Dysbiosis. Curr Nutr Rep 2025; 14:25. [PMID: 39890659 DOI: 10.1007/s13668-025-00614-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE OF REVIEW The gut microbiota (GM) is directly related to health and disease. In this context, disturbances resulting from excessive stress, unbalanced diet, alcohol abuse, and antibiotic use, among other factors, can contribute to microbiota imbalance, with significant impacts on host health. This review provides a comprehensive examination of the literature on the influence of diet on dysbiosis and increased intestinal permeability over the past five years. RECENT FINDINGS Diet can be considered one of the main modulating factors of GM, impacting its composition and functionality. Excessive consumption of simple carbohydrates, saturated fats, and processed foods appears to be directly linked to dysbiosis, which can lead to intestinal hyperpermeability and leaky gut syndrome. On the other hand, diets primarily composed of food groups such as nuts, vegetables, fruits, fish, and poultry in moderate quantities, along with limited consumption of red and processed meats, are associated with a more diverse, healthier, and beneficial GM for the host. It is worth noticing that the use of prebiotics and probiotics, omega-3 supplementation, polyunsaturated fatty acids, and vitamins A, B, C, D, and E can positively modulate the intestinal microbiota by altering its metabolic activity, microbial composition, and improve intestinal barrier function. This review points to a new perspective regarding individualized dietary intervention and the need to integrate it into several aspects of cellular biology, biochemistry, and microbiology to prescribe more effective diets and thus contribute to patients' comprehensive health.
Collapse
Affiliation(s)
- Ana Beatriz Marcari
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Claudio Roberto Simon
- Department of Structural Biology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil
| | - Maria Emilia Soares Martins Dos Santos
- Department of Biochemistry, Pharmacology and Physiology, Federal University of Triângulo Mineiro, Praça Manoel Terra, 330 - Abadia, Uberaba, MG, 38025-015, Brazil.
| |
Collapse
|
5
|
Zeng Z, Gong S, Quan C, Zhou S, Kulyar MFEA, Iqbal M, Li Y, Li X, Li J. Impact of Bacillus licheniformis from yaks following antibiotic therapy in mouse model. Appl Microbiol Biotechnol 2024; 108:139. [PMID: 38229401 DOI: 10.1007/s00253-023-12866-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: 06/27/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 01/18/2024]
Abstract
Gut microorganism (GM) is an integral component of the host microbiome and health system. Abuse of antibiotics disrupts the equilibrium of the microbiome, affecting environmental pathogens and host-associated bacteria alike. However, relatively little research on Bacillus licheniformis alleviates the adverse effects of antibiotics. To test the effect of B. licheniformis as a probiotic supplement against the effects of antibiotics, cefalexin was applied, and the recovery from cefalexin-induced jejunal community disorder and intestinal barrier damage was investigated by pathology, real-time PCR (RT-PCR), and high-throughput sequencing (HTS). The result showed that A group (antibiotic treatment) significantly reduced body weight and decreased the length of jejunal intestinal villi and the villi to crypt (V/C) value, which also caused structural damage to the jejunal mucosa. Meanwhile, antibiotic treatment suppressed the mRNA expression of tight junction proteins ZO-1, claudin, occludin, and Ki67 and elevated MUC2 expression more than the other Groups (P < 0.05 and P < 0.01). However, T group (B. licheniformis supplements after antibiotic treatment) restored the expression of the above genes, and there was no statistically significant difference compared to the control group (P > 0.05). Moreover, the antibiotic treatment increased the relative abundance of 4 bacterial phyla affiliated with 16 bacterial genera in the jejunum community, including the dominant Firmicutes, Proteobacteria, and Cyanobacteria in the jejunum. B. licheniformis supplements after antibiotic treatment reduced the relative abundance of Bacteroidetes and Proteobacteria and increased the relative abundance of Firmicutes, Epsilonbacteraeota, Lactobacillus, and Candidatus Stoquefichus. This study uses mimic real-world exposure scenarios by considering the concentration and duration of exposure relevant to environmental antibiotic contamination levels. We described the post-antibiotic treatment with B. licheniformis could restore intestinal microbiome disorders and repair the intestinal barrier. KEY POINTS: • B. licheniformis post-antibiotics restore gut balance, repair barrier, and aid health • Antibiotics harm the gut barrier, alter structure, and raise disease risk • Long-term antibiotics affect the gut and increase disease susceptibility.
Collapse
Affiliation(s)
- Zhibo Zeng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Saisai Gong
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chuxian Quan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shimeng Zhou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | | | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Yan Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiang Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
- College of Animals Husbandry and Veterinary Medicine, Tibet Agricultural and Animal Husbandry University, Linzhi, 860000, Tibet, China.
| |
Collapse
|
6
|
Ughade PA, Shrivastava D, Chaudhari K. Navigating the Microbial Landscape: Understanding Dysbiosis in Human Genital Tracts and Its Impact on Fertility. Cureus 2024; 16:e67040. [PMID: 39286717 PMCID: PMC11403153 DOI: 10.7759/cureus.67040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Dysbiosis, an imbalance in microbial communities, significantly impacts the health and functionality of the human genital tract, with profound implications for fertility and reproductive health. This review explores the intricate relationship between genital tract microbiota and reproductive outcomes, highlighting the composition and dynamics of these microbial communities in both females and males. In females, the vaginal microbiota, primarily dominated by Lactobacillus species, is essential for maintaining a healthy vaginal environment, preventing infections, and supporting reproductive functions. In males, the genital microbiota influences sperm quality and reproductive health. Dysbiosis in the genital tract, manifesting as bacterial vaginosis, yeast infections, urethritis, or prostatitis, disrupts these microbial communities, leading to adverse reproductive outcomes such as infertility, pregnancy, and increased susceptibility to sexually transmitted infections. This review delves into the mechanisms through which dysbiosis affects fertility, including alterations in vaginal pH, mucosal immunity, inflammation, sperm viability, and motility. It also evaluates diagnostic methods, clinical implications, and management strategies, including probiotics, prebiotics, antibiotics, antifungal treatments, lifestyle interventions, and emerging therapeutic approaches. By understanding the microbial landscape of the genital tract and its impact on fertility, this review aims to inform targeted interventions that restore microbial balance and enhance reproductive health, ultimately improving fertility outcomes and the potential for healthy pregnancies.
Collapse
Affiliation(s)
- Prachi A Ughade
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Deepti Shrivastava
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Kamlesh Chaudhari
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
7
|
Duan R, Liu Y, Zhang Y, Shi J, Xue R, Liu R, Miao Y, Zhou X, Lv Y, Shen H, Xie X, Ai X. The impact of exercise on the gut microbiota in middle-aged amateur serious runners: a comparative study. Front Physiol 2024; 15:1343219. [PMID: 38737829 PMCID: PMC11082653 DOI: 10.3389/fphys.2024.1343219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Exercise, health, and the gut microbiota (GM) are strongly correlated. Research indicates that professional athletes, especially ultra-marathon runners, have unique GM characteristics. However, more research has focused on elite athletes, with little attention given to amateur sports enthusiasts, especially those in the middle-aged population. Therefore, this study focuses on the impact of long-term running on the composition and potential functions of the GM in middle-aged individuals. Methods We compared the GM of 25 middle-aged serious runnerswith 22 sedentary healthy controls who had minimal exercise habitsusing 16S rRNA gene sequencing. Additionally, we assessed dietary habits using a food frequency questionnaire. Results and Discussion Statistical analysis indicates that there is no significant difference in dietary patterns between the control group and serious runners. Diversity analysis results indicate that there is no significant difference in α diversity between the two groups of GM, but there is a significant difference in β diversity. Analysis of the composition of GM reveals that Ruminococcus and Coprococcus are significantly enriched in serious runners, whereas Bacteroides, Lachnoclostridium, and Lachnospira are enriched in the control group. Differential analysis of functional pathway prediction results reveals significant differences in the functional metabolism levels of GM between serious runners and the control group. Further correlation analysis results indicate that this difference may be closely related to variations in GM. In conclusion, our results suggest that long-term exercise can lead to changes in the composition of the GM. These changes have the potential to impact the overall health of the individual by influencing metabolic regulation.
Collapse
Affiliation(s)
- Rui Duan
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Yu Liu
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Yonglian Zhang
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Jinrong Shi
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Rong Xue
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Ruijie Liu
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Yuanxin Miao
- Research Institute of Agricultural Biotechnology, Jingchu University of Technology, Jingmen, Hubei, China
| | - Xianfeng Zhou
- School of Life Sciences and Health Engineering, Hubei University of Technology, Wuhan, China
- Maintainbiotech Ltd., Wuhan, Hubei, China
| | | | - Hexiao Shen
- Maintainbiotech Ltd., Wuhan, Hubei, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiongwei Xie
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| | - Xu Ai
- Jingmen Central Hospital, Hubei Clinical Medical Research Center for Functional Colorectal Diseases, Jingmen, Hubei, China
| |
Collapse
|
8
|
Chow EPF, Fairley CK, Kong FYS. STI pathogens in the oropharynx: update on screening and treatment. Curr Opin Infect Dis 2024; 37:35-45. [PMID: 38112085 DOI: 10.1097/qco.0000000000000997] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
PURPOSE OF REVIEW The rise in antimicrobial resistance in several STI pathogens such as Neisseria gonorrhoeae has become a public health threat as only one first-line treatment remains. Reducing screening interval for gonorrhoea and chlamydia in high-prevalence populations has been proposed to address antimicrobial stewardship, but this remains controversial. This review aimed to revisit the epidemiology of infections at the oropharynx and review the current screening recommendations and treatment guidelines in different populations. RECENT FINDINGS Emerging evidence suggests that the oropharynx is the primary anatomical site for gonorrhoea transmission but maybe not for chlamydia transmission. Most international guidelines recommend 3-monthly oropharyngeal gonorrhoea and chlamydia screening for high-prevalence populations (e.g. men who have sex with men) but not low-prevalence populations (e.g. heterosexuals) given the clinical and public health benefits of screening in low-prevalence populations are still unclear. Doxycycline remains the first-line treatment for oropharyngeal chlamydia in most guidelines. However, some countries have moved from dual therapy (ceftriaxone and azithromycin) to monotherapy (ceftriaxone) for oropharyngeal gonorrhoea treatment to address antimicrobial stewardship. SUMMARY The transmission of gonorrhoea and chlamydia is still not fully understood. Further work will be required to evaluate the benefits and harms of reducing screening in high-prevalence populations.
Collapse
Affiliation(s)
- Eric P F Chow
- Melbourne Sexual Health Centre, Alfred Health
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Christopher K Fairley
- Melbourne Sexual Health Centre, Alfred Health
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University
| | - Fabian Y S Kong
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
9
|
Burz SD, Causevic S, Dal Co A, Dmitrijeva M, Engel P, Garrido-Sanz D, Greub G, Hapfelmeier S, Hardt WD, Hatzimanikatis V, Heiman CM, Herzog MKM, Hockenberry A, Keel C, Keppler A, Lee SJ, Luneau J, Malfertheiner L, Mitri S, Ngyuen B, Oftadeh O, Pacheco AR, Peaudecerf F, Resch G, Ruscheweyh HJ, Sahin A, Sanders IR, Slack E, Sunagawa S, Tackmann J, Tecon R, Ugolini GS, Vacheron J, van der Meer JR, Vayena E, Vonaesch P, Vorholt JA. From microbiome composition to functional engineering, one step at a time. Microbiol Mol Biol Rev 2023; 87:e0006323. [PMID: 37947420 PMCID: PMC10732080 DOI: 10.1128/mmbr.00063-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
Abstract
SUMMARYCommunities of microorganisms (microbiota) are present in all habitats on Earth and are relevant for agriculture, health, and climate. Deciphering the mechanisms that determine microbiota dynamics and functioning within the context of their respective environments or hosts (the microbiomes) is crucially important. However, the sheer taxonomic, metabolic, functional, and spatial complexity of most microbiomes poses substantial challenges to advancing our knowledge of these mechanisms. While nucleic acid sequencing technologies can chart microbiota composition with high precision, we mostly lack information about the functional roles and interactions of each strain present in a given microbiome. This limits our ability to predict microbiome function in natural habitats and, in the case of dysfunction or dysbiosis, to redirect microbiomes onto stable paths. Here, we will discuss a systematic approach (dubbed the N+1/N-1 concept) to enable step-by-step dissection of microbiome assembly and functioning, as well as intervention procedures to introduce or eliminate one particular microbial strain at a time. The N+1/N-1 concept is informed by natural invasion events and selects culturable, genetically accessible microbes with well-annotated genomes to chart their proliferation or decline within defined synthetic and/or complex natural microbiota. This approach enables harnessing classical microbiological and diversity approaches, as well as omics tools and mathematical modeling to decipher the mechanisms underlying N+1/N-1 microbiota outcomes. Application of this concept further provides stepping stones and benchmarks for microbiome structure and function analyses and more complex microbiome intervention strategies.
Collapse
Affiliation(s)
- Sebastian Dan Burz
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Senka Causevic
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Alma Dal Co
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Marija Dmitrijeva
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Philipp Engel
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Daniel Garrido-Sanz
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Gilbert Greub
- Institut de microbiologie, CHUV University Hospital Lausanne, Lausanne, Switzerland
| | | | | | | | - Clara Margot Heiman
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | | | - Christoph Keel
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - Soon-Jae Lee
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Julien Luneau
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Lukas Malfertheiner
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Sara Mitri
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Bidong Ngyuen
- Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Omid Oftadeh
- Laboratory of Computational Systems Biotechnology, EPF Lausanne, Lausanne, Switzerland
| | | | | | - Grégory Resch
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, CHUV University Hospital Lausanne, Lausanne, Switzerland
| | | | - Asli Sahin
- Laboratory of Computational Systems Biotechnology, EPF Lausanne, Lausanne, Switzerland
| | - Ian R. Sanders
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | | | - Janko Tackmann
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Robin Tecon
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - Jordan Vacheron
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - Evangelia Vayena
- Laboratory of Computational Systems Biotechnology, EPF Lausanne, Lausanne, Switzerland
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
10
|
Kenyon C, Herrmann B, Hughes G, de Vries HJ. Management of asymptomatic sexually transmitted infections in Europe: towards a differentiated, evidence-based approach. THE LANCET REGIONAL HEALTH. EUROPE 2023; 34:100743. [PMID: 37927435 PMCID: PMC10624996 DOI: 10.1016/j.lanepe.2023.100743] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/05/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023]
Abstract
Most sexually transmitted infections (STIs) can be accurately diagnosed and treated during asymptomatic carriage. Widespread screening for these STIs is therefore assumed to be an effective way to reduce their prevalence and associated disease. In this review, we provide evidence that this is the case for HIV and syphilis. However, for other STIs such as Neisseria gonorrhoeae and Chlamydia trachomatis, our review reveals that the evidence that screening reduces infection prevalence and associated disease is weak. There is also growing evidence of harms from screening that might outweigh any benefits. The harms include the increased consumption of antimicrobials that follows frequent screening and increased detection of asymptomatic STIs in key populations, such as men who have sex with men taking HIV pre-exposure prophylaxis, and associated risk of antimicrobial resistance in target and non-target organisms. There may also be psycho-social harm associated with an STI diagnosis. We conclude that in the absence of symptoms, in high STI prevalence populations frequent STI screening should be limited to HIV and syphilis.
Collapse
Affiliation(s)
- Chris Kenyon
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Björn Herrmann
- Section of Clinical Microbiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gwenda Hughes
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, UK
| | - Henry J.C. de Vries
- Department of Dermatology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
- Center for Sexual Health, Department of Infectious Diseases, Public Health Service Amsterdam, the Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Du M, Ren Z, Li Q, Pu Q, Li X, Qiu Y, Li Y. Reduced bacterial resistance antibiotics with improved microbiota tolerance in human intestinal: Molecular design and mechanism analysis. JOURNAL OF HAZARDOUS MATERIALS 2023; 460:132368. [PMID: 37619278 DOI: 10.1016/j.jhazmat.2023.132368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
Antibiotic selectivity and bacterial resistance are critical global public health issues. We constructed a multi-class machine learning model to study antibiotic effects on human intestinal microbiota abundance and identified key features. Binding energies of β-lactam antibiotics with Escherichia coli PBP3 mutant protein were calculated, and a 2D-QSAR model for bacterial resistance was established. Sensitivity analysis identified key features affecting bacterial resistance. By coupling key features from the machine learning model and 2D-QSAR model, we designed ten flucloxacillin (FLU) substitutes that improved intestinal microbiota tolerance and reduced antibiotic bacterial resistance. Concurrently, the substitutes exhibited superior degradability in soil, aquatic environments, and under photolytic conditions, coupled with a reduced environmental toxicity compared to the FLU. Evaluations under combined medication revealed significant improvements in functionality and bacterial resistance for 80% of FLU substitutes, with 50% showing more than a twofold increase. Mechanistic analysis demonstrated enhanced binding to target proteins and increased biodegradability for FLU substitutes due to more concentrated surface charges. Reduced solvent hindrance and increased cell membrane permeability of FLU substitutes, mainly due to enhanced interactions with phospholipid bilayers, contributed to their functional selectivity. This study aims to address poor antibiotic selectivity and strong bacterial resistance, providing guidance for designing antibiotic substitutes.
Collapse
Affiliation(s)
- Meijin Du
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Zhixing Ren
- College of Forestry, Northeast Forestry University, Harbin 150040, China
| | - Qing Li
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Qikun Pu
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Xinao Li
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Youli Qiu
- School of Chemical Safety, North China Institute of Science and Technology, Yanjiao 065201, China.
| | - Yu Li
- College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| |
Collapse
|
12
|
Dela Cruz M, Lin H, Han J, Adler E, Boissiere J, Khalid M, Sidebottom A, Sundararajan A, Lehmann C, Moran A, Odenwald M, Stutz M, Kim G, Pinney S, Jeevanandam V, Alegre ML, Pamer E, Nguyen AB. Reduced immunomodulatory metabolite concentrations in peri-transplant fecal samples from heart allograft recipients. FRONTIERS IN TRANSPLANTATION 2023; 2:1182534. [PMID: 38993864 PMCID: PMC11235359 DOI: 10.3389/frtra.2023.1182534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2024]
Abstract
Background Emerging evidence is revealing the impact of the gut microbiome on hematopoietic and solid organ transplantation. Prior studies postulate that this influence is mediated by bioactive metabolites produced by gut-dwelling commensal bacteria. However, gut microbial metabolite production has not previously been measured among heart transplant (HT) recipients. Methods In order to investigate the potential influence of the gut microbiome and its metabolites on HT, we analyzed the composition and metabolite production of the fecal microbiome among 48 HT recipients at the time of HT. Results Compared to 20 healthy donors, HT recipients have significantly reduced alpha, i.e. within-sample, microbiota diversity, with significantly lower abundances of key anaerobic commensal bacteria and higher abundances of potentially pathogenic taxa that have been correlated with adverse outcomes in other forms of transplantation. HT recipients have a wide range of microbiota-derived fecal metabolite concentrations, with significantly reduced levels of immune modulatory metabolites such as short chain fatty acids and secondary bile acids compared to healthy donors. These differences were likely due to disease severity and prior antibiotic exposures but were not explained by other demographic or clinical factors. Conclusions Key potentially immune modulatory gut microbial metabolites are quantifiable and significantly reduced among HT recipients compared to healthy donors. Further study is needed to understand whether this wide range of gut microbial dysbiosis and metabolite alterations impact clinical outcomes and if they can be used as predictive biomarkers or manipulated to improve transplant outcomes.
Collapse
Affiliation(s)
- Mark Dela Cruz
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Jiho Han
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Emerald Adler
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Jaye Boissiere
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Maryam Khalid
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Ashley Sidebottom
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Anitha Sundararajan
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Christopher Lehmann
- Department of Medicine, Section of Infectious Diseases, University of Chicago Medicine, Chicago, IL, United States
| | - Angelica Moran
- Department of Pathology, University of Chicago Medicine, Chicago, IL, United States
| | - Matthew Odenwald
- Department of Medicine, Section of Gastroenterology, University of Chicago Medicine, Chicago, IL, United States
| | - Matthew Stutz
- Department of Medicine, Section of Pulmonary and Critical Care, University of Chicago Medicine, Chicago, IL, United States
| | - Gene Kim
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Sean Pinney
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| | - Valluvan Jeevanandam
- Department of Surgery, Section of Cardiac Surgery, University of Chicago Medicine, Chicago, IL, United States
| | - Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, University of Chicago, Chicago, IL, United States
| | - Eric Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, United States
| | - Ann B. Nguyen
- Department of Medicine, Section of Cardiology, University of Chicago Medicine, Chicago, IL, United States
| |
Collapse
|
13
|
Pecyna P, Gabryel M, Mankowska-Wierzbicka D, Nowak-Malczewska DM, Jaskiewicz K, Jaworska MM, Tomczak H, Rydzanicz M, Ploski R, Grzymislawski M, Dobrowolska A, Gajecka M. Gender Influences Gut Microbiota among Patients with Irritable Bowel Syndrome. Int J Mol Sci 2023; 24:10424. [PMID: 37445604 DOI: 10.3390/ijms241310424] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic functional gastrointestinal disease that affects approximately 11% of the general population. The gut microbiota, among other known factors, plays a substantial role in its pathogenesis. The study aimed to characterize the gut microbiota differences between patients with IBS and unaffected individuals, taking into account the gender aspect of the patients and the types of IBS determined on the basis of the Rome IV Criteria, the IBS-C, IBS-D, IBS-M, and IBS-U. In total, 121 patients with IBS and 70 unaffected individuals participated in the study; the derived stool samples were subjected to 16S rRNA amplicon sequencing. The gut microbiota of patients with IBS was found to be more diverse in comparison to unaffected individuals, and the differences were observed primarily among Clostridiales, Mogibacteriaceae, Synergistaceae, Coriobacteriaceae, Blautia spp., and Shuttleworthia spp., depending on the study subgroup and patient gender. There was higher differentiation of females' gut microbiota compared to males, regardless of the disease status. No correlation between the composition of the gut microbiota and the type of IBS was found. Patients with IBS were characterized by more diverse gut microbiota compared to unaffected individuals. The gender criterion should be considered in the characterization of the gut microbiota. The type of IBS did not determine the identified differences in gut microbiota.
Collapse
Affiliation(s)
- Paulina Pecyna
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Marcin Gabryel
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Dorota Mankowska-Wierzbicka
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Dorota M Nowak-Malczewska
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | | | - Marcelina M Jaworska
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Hanna Tomczak
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- Central Microbiology Laboratory, H. Swiecicki Clinical Hospital at the Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Malgorzata Rydzanicz
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland
| | - Rafal Ploski
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland
| | - Marian Grzymislawski
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Agnieszka Dobrowolska
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Marzena Gajecka
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
| |
Collapse
|
14
|
Alverdy JC. Rationale for Colonic Pre-Habilitation Prior to Restoration of Gastrointestinal Continuity. Surg Infect (Larchmt) 2023; 24:265-270. [PMID: 37010975 PMCID: PMC10061335 DOI: 10.1089/sur.2023.001] [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] [Indexed: 04/04/2023] Open
Abstract
The emergence of the gut microbiome as a complex ecosystem that plays a key role in human heath and disease has touched virtually every aspect of medical and surgical care. With the advent of next-generation technology to interrogate the microbiome at the level of its membership, community structure and production of metabolites, applying measures by which the gut microbiome can be manipulated to the advantage of both the patient and provider is now possible. Among the many proposed methods, the most practical and promising is dietary pre-habilitation of the gut microbiome prior to high-risk anastomotic surgery. In this review, we will outline the scientific rationale and molecular underpinning that support dietary pre-habilitation as a practical and deliverable method to prevent complications after high-risk anastomotic surgery.
Collapse
Affiliation(s)
- John C. Alverdy
- Department of Surgery, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
15
|
Vázquez-Cabrera N, Espinosa-Márquez A, Cedillo-Ramírez ML. [Historical evolution of World Health Organization guidelines on antimicrobial resistanceEvolução histórica da Organização Mundial da Saúde e a resistência aos antimicrobianos]. Rev Panam Salud Publica 2023; 47:e51. [PMID: 36843661 PMCID: PMC9943709 DOI: 10.26633/rpsp.2023.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/15/2022] [Indexed: 02/25/2023] Open
Abstract
Objective Show the evolution of guidelines on public health policies focused on antimicrobial resistance (AMR) issued by the World Health Organization (WHO) between 1948 and 2022. Other related government actions are also mentioned. Methods A detailed review was conducted of World Health Assembly and WHO Executive Board archives. A textual analysis was conducted of AMR-related resolutions that guide the design of government policies and actions for WHO Member States. A systematic search was carried out in SCOPUS, PubMed, and grey literature under the category of public health policies on AMR. Results AMR has become the greatest threat to public health, putting at risk the achievement of the Sustainable Development Goals. WHO resolutions are presented as evidence of guidelines to combat AMR. The One Health approach and related strategies, initiatives, plans, and programs are mentioned. A gap was identified in the research and development of new antimicrobials, requiring further analysis. Conclusions WHO has made efforts to combat AMR. This has generated comprehensive development of public health policies to be implemented by the governments of Member States as they see fit.
Collapse
Affiliation(s)
- Nixache Vázquez-Cabrera
- Instituto de CienciasBenemérita Universidad Autónoma de PueblaMéxicoInstituto de Ciencias, Benemérita Universidad Autónoma de Puebla, México.
| | - Araceli Espinosa-Márquez
- Instituto de Ciencias de Gobierno y Desarrollo EstratégicoBenemérita Universidad Autónoma de PueblaMéxicoInstituto de Ciencias de Gobierno y Desarrollo Estratégico, Benemérita Universidad Autónoma de Puebla, México.
| | - María Lilia Cedillo-Ramírez
- Centro de Detección BiomolecularBenemérita Universidad Autónoma de PueblaMéxicoCentro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, México.
| |
Collapse
|
16
|
Dvořák Z, Li H, Mani S. Microbial Metabolites as Ligands to Xenobiotic Receptors: Chemical Mimicry as Potential Drugs of the Future. Drug Metab Dispos 2023; 51:219-227. [PMID: 36184080 PMCID: PMC9900867 DOI: 10.1124/dmd.122.000860] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/28/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023] Open
Abstract
Xenobiotic receptors, such as the pregnane X receptor, regulate multiple host physiologic pathways including xenobiotic metabolism, certain aspects of cellular metabolism, and innate immunity. These ligand-dependent nuclear factors regulate gene expression via genomic recognition of specific promoters and transcriptional activation of the gene. Natural or endogenous ligands are not commonly associated with this class of receptors; however, since these receptors are expressed in a cell-type specific manner in the liver and intestines, there has been significant recent effort to characterize microbially derived metabolites as ligands for these receptors. In general, these metabolites are thought to be weak micromolar affinity ligands. This journal anniversary minireview focuses on recent efforts to derive potentially nontoxic microbial metabolite chemical mimics that could one day be developed as drugs combating xenobiotic receptor-modifying pathophysiology. The review will include our perspective on the field and recommend certain directions for future research. SIGNIFICANCE STATEMENT: Xenobiotic receptors (XRs) regulate host drug metabolism, cellular metabolism, and immunity. Their presence in host intestines allows them to function not only as xenosensors but also as a response to the complex metabolic environment present in the intestines. Specifically, this review focuses on describing microbial metabolite-XR interactions and the translation of these findings toward discovery of novel chemical mimics as potential drugs of the future for diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Zdeněk Dvořák
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Hao Li
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Departments of Medicine (H.L., S.M.), Molecular Pharmacology (S.M.), and Genetics (S.M.), Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
17
|
Kumari R, Singhvi N, Sharma P, Choudhury C, Shakya R. Virtual screening of gut microbiome bacteriocins as potential inhibitors of stearoyl-CoA desaturase 1 to regulate adipocyte differentiation and thermogenesis to combat obesity. J Biomol Struct Dyn 2023; 41:12632-12642. [PMID: 36644882 DOI: 10.1080/07391102.2023.2167121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
The gut bacterial strains and their metabolites have been shown to play a significant role in obesity, but the molecular mechanisms underlying this association are largely unresolved. Obesity is a multifactorial problem and is controlled by various mechanisms and pathways to produce and store fat cells. Bacteriocins are secondary metabolites produced by gut bacteria to defend themselves against their competitors. Recently, they have gained great attention due to their role in metabolic disorders, including obesity. Stearoyl-CoA desaturase 1 (SCD1) is a key enzyme involved in the differentiation of adipocytes. The aim of this study is to show the regulation of SCD1 by bacteriocins and thus their importance in obesity control. We screened the human gut bacteriome for the presence of bacteriocins, predicted their structures, and showed their inhibitory role by molecular docking with SCD1. Further, to confirm the docking results, MDS of six top scoring SCD1-bacteriocin complexes were carried out for 100 ns. These six bacteriocins namely, Plantaricin S-beta, Carnolysin, Lactococcin B, Bacteriocin Iic, Plantaricin N, and Thermophilin A, with strong binding affinities, are primarily produced by bacterial strains from the Lactobacillaeacea family. These findings can be the basis of further experiments for enhanced understanding of the underlying mechanisms for obesity control, specifically bacteriocins driven regulation of the SCD1 enzyme. In addition, a consortium of bacterial strains producing these bacteriocins can be developed and used as probiotics for the amelioration of obesity and other metabolic complications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rekha Kumari
- Molecular Microbiology and Bioinformatics Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Nirjara Singhvi
- School of Allied Sciences, Dev Bhoomi Uttarakhand University, Dehradun, Uttarakhand, India
| | - Poonam Sharma
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Chinmayee Choudhury
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Rashmi Shakya
- Department of Botany, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|