1
|
Lin Z, Zhang P, Shi D, Zhang Y, Wu W, Tang Q, Wang Q, Wang S. Association between the gut microbiota and cystitis: A two-sample mendelian randomization study combined with the GEO database. Microb Pathog 2025:107683. [PMID: 40348210 DOI: 10.1016/j.micpath.2025.107683] [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: 09/22/2024] [Revised: 04/26/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Disturbances within the intestinal microbiota have emerged as a significant factor contributing to systemic inflammation, thereby rendering distant anatomical sites more vulnerable to various illnesses, including inflammatory conditions in the urinary tract such as cystitis. However, the causal relationship between dysbiosis of the gut microbiota and cystitis remains unclear. We sought to elucidate the causal relationship between the intestinal microbiota and cystitis employing Mendelian randomization (MR), offering insights into novel preventive and therapeutic strategies for managing cystitis. METHOD Summary statistics for the Genome-Wide Association Study (GWAS) of cystitis were sourced from the R5 release dataset provided by the FinnGen consortium, which included 8,081 cystitis cases and 195,140 controls. Single Nucleotide Polymorphisms (SNPs) that showed strong associations with 196 microbial taxa (encompassing 18,340 individuals) were selected as instrumental variables. To analyse the causal relationships between cystitis and gut microbiota, we employed four MR analysis methods: random effects, inverse variance weighting, weighted medians, and MR-Egger regression. Sensitivity analyses were performed using the Cochran's Q test, funnel plots, leave-one-out analyses, and the MR-Egger intercept test. We conducted metagenomic analysis of fecal samples from 7 patients with cystitis and 7 healthy controls to validate the findings from our MR results. To further elucidate the biological mechanisms, we conducted positional mapping of the extracted SNPs associated with the significant taxa. Additionally, we curated differentially expressed genes (DEGs) from three datasets about cystitis obtained from the Gene Expression Omnibus (GEO). Finally, we intersected the DEGs with the mapped genes to identify common genes of cystitis. RESULTS Our analysis revealed significant associations between specific gut microbiota and cystitis. IVW results revealed that four gut microorganisms, specifically, the genus RuminococcaceaeUCG011, genus Sutterella, family Porphyromonadaceae, and family Veillonellaceae (P < 0.05), contributed to a reduction in the incidence of cystitis. Similarly, four cystitis-related bacteria, namely, the genus Marvinbryantia, the genus Odoribacter, the genus Ruminiclostridium6, and the genus Sellimonas, are thought to play a significant role in elevating the risk of cystitis (P < 0.05). The metagenomic analysis revealed significant differences in the abundance of the genera Sutterella and Odoribacter in patients with cystitis compared to healthy controls. Additionally, we mapped causal SNPs to genes and identified 62 genes. Bioinformatics analysis reveals 161 common DEGs in cystitis. Through MR and bioinformatics analysis, we identified two common genes-ICAM1 and HP-as potential targets for cystitis. CONCLUSION Our research identified genetic connections between eight components of gut microbiota and two genes related to cystitis. These results offer important insights for subsequent studies into the complex relationship between gut microbiota and cystitis.
Collapse
Affiliation(s)
- Zeyu Lin
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ping Zhang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Donghui Shi
- Department of Urology, Suzhou Wu Zhong People's Hospital, Suzhou, 215100, China
| | - Yuyan Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wenqiang Wu
- Department of Urology, Chongqing Hospital of Jiangsu Province Hospital, Chongqing 401420, China
| | - Qingsheng Tang
- Department of Urology, People's Hospital of Dongtai City, Dongtai, Jiangsu, China.
| | - Qing Wang
- Department of Urology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China.
| | - Shangqian Wang
- Department of Urology, Chongqing Hospital of Jiangsu Province Hospital, Chongqing 401420, China; Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China; State Key Laboratory of Reproductive Medicine and offspring health, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
2
|
Münch A, Escudero-Hernández C. Dissecting Microscopic Colitis Immunopathophysiology: Insights From Basic Research. United European Gastroenterol J 2025. [PMID: 40317876 DOI: 10.1002/ueg2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 05/07/2025] Open
Abstract
Microscopic colitis is an inflammatory bowel disease (IBD) comprising two clinically undiscernible entities: collagenous colitis and lymphocytic colitis. Collagenous colitis associates with HLA genes and displays a Th1/Tc1-Th17/Tc17 profile with pericryptal myofibroblast activity, water malabsorption and secondary fluid loss due to altered osmoregulation. Conversely, lymphocytic colitis lacks genetic associations and displays a Th1/Th2 profile and paracellular/transcellular permeability. Lymphocytic colitis subclassifies into channelopathic lymphocytic colitis due to unique alteration of ion and organic acid transport that could result from drug exposure, and inflammatory lymphocytic colitis due to the involvement of moderate immune responses compared to collagenous colitis. As microscopic colitis mucosa remains intact and immune cells seem to stay inactive, microscopic colitis is an ideal model to explore early stages of IBD if collagenous colitis and lymphocytic colitis are studied as distinct entities. Exploiting multiomic approaches and established biobanks will ensure validation of microscopic colitis patient stratification, and deepening into pathomechanisms which could enable precision medicine.
Collapse
Affiliation(s)
- Andreas Münch
- Department of Gastroenterology and Hepatology, Linköping University, Linköping, Sweden
- Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Celia Escudero-Hernández
- Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
3
|
Nichols L, El-Kholy O, Elsayed AAR, Basson MD. The bidirectional interplay between gut dysbiosis and surgical complications: A systematic review. Am J Surg 2025; 245:116369. [PMID: 40344995 DOI: 10.1016/j.amjsurg.2025.116369] [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: 03/30/2025] [Revised: 04/16/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The gut microbiome critically influences diverse aspects of physiology and surgical recovery. Conversely, surgery alters the microbiome, potentially predisposing to complications. We aimed to clarify the bidirectional interaction between surgery and gut dysbiosis. METHODS On December 22nd, 2024, a systematic search of the Cochrane Library, PubMed, VHL, and WOS was completed. Relevant studies were assessed for risk of bias using STROBE and CONSORT guidelines. RESULTS Thirty studies, with 2500+ participants experiencing diverse procedures and complications, were incorporated. Although specifics varied, dysbiosis correlated with surgery and its complications. Patients with complications had more harmful bacteria and fewer beneficial bacteria. In some studies, probiotics reduced complications. CONCLUSION Gut dysbiosis is tied to postoperative complications in a complex, bidirectional relationship. Patients with surgical complications may have fewer beneficial and more pathogenic bacteria both before and after surgery. Early identification of dysbiosis and probiotic administration could predict or even reduce postoperative complications.
Collapse
Affiliation(s)
- Lindsey Nichols
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Omar El-Kholy
- Faculty of Medicine, Alexandria University, Alexandria, 21521, Egypt
| | - Ahmed Adham R Elsayed
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Surgery, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Marc D Basson
- College of Medicine, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Surgery, Northeast Ohio Medical University, Rootstown, OH, 44272, USA; Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, 44272, USA.
| |
Collapse
|
4
|
Sørum ME, Boulund U, De Pietri S, Weischendorff S, Enevold C, Rathe M, Als-Nielsen B, Hasle H, Pamp S, Stokholm J, Müller K. Changes in gut microbiota predict neutropenia after induction treatment in childhood acute lymphoblastic leukemia. Blood Adv 2025; 9:1508-1521. [PMID: 39561377 PMCID: PMC11985026 DOI: 10.1182/bloodadvances.2024013986] [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: 06/14/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Delayed neutrophil recovery during acute lymphoblastic leukemia (ALL) treatment increases the risk of infection and causes delay in chemotherapy. Emerging evidence implicates gut microbiota in neutrophil reconstitution after chemotherapy. We explored the interplay between the gut microbiota and neutrophil dynamics, including neutrophil chemoattractants, in 51 children with newly diagnosed ALL. Daily absolute neutrophil count (ANC), weekly plasma chemokines (CXCL1 and CXCL8), granulocyte colony-stimulating factor (G-CSF), and fecal samples were monitored until day 29 during ALL induction treatment. Fecal sequencing using 16S ribosomal RNA revealed an overall significant reduction in bacterial diversity and Enterococcus overgrowth throughout the induction treatment. Prolonged neutropenia (ANC <0.5 × 109 cells per L at day 36) and elevated chemokine levels were associated with a decreased abundance of genera from the Ruminococcaceae and Lachnospiraceae families, decreased Veillonella genus, and Enterococcus overgrowth from diagnosis and throughout induction treatment. G-CSF was upregulated in response to neutropenia but was unrelated to microbiota changes. Overall, this study revealed that a diminished abundance of specific intestinal commensals and Enterococcus overgrowth is associated with delayed neutrophil reconstitution and increased chemokine signaling, indicating that disruption of the microbiota may contribute to prolonged neutropenia. These findings lay the groundwork for future investigations into the mechanisms underlying these associations and their clinical implications for developing gut-sparring strategies to minimize the impact of gut dysbiosis on immune recovery.
Collapse
Affiliation(s)
- Maria Ebbesen Sørum
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Silvia De Pietri
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Weischendorff
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mathias Rathe
- Department of Pediatrics and Adolescent Medicine, The Hans Christian Andersen Children’s Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Sünje Pamp
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Freitas JA, Nehmi Filho V, Santamarina AB, Murata GM, Franco LAM, Fonseca JV, Martins RC, Souza GA, Benicio G, Sabbag IM, de Souza EA, Otoch JP, Pessoa AFM. Nutraceutical supplement slim reshaped colon histomorphology and reduces Mucispirillum schaedleri in obese mice. Front Microbiol 2025; 16:1494994. [PMID: 40236479 PMCID: PMC11997693 DOI: 10.3389/fmicb.2025.1494994] [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: 09/24/2024] [Accepted: 02/28/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction Bioactive compounds and whole foods have emerged as promising interventions to address gut microbiota dysbiosis linked to obesity. Compounds such as berberine and coenzyme Q10 are well-recognized for their roles in managing metabolic syndrome and exerting antioxidant effects, while beet pulp, rich in fiber and antioxidants, enhances gut health through additional prebiotic benefits. Methods This study evaluated the effects of a nutraceutical supplement, Slim, on the modulation of gut microbiota in obese mice induced by a high-fat diet. Results Our results demonstrated that Slim supplementation significantly improved lipid metabolism, reshaped colon histomorphology, and decreased levels of Mucispirillum schaedleri, which were correlated with VLDL-c and triglycerides. Discussion We suggest these effects are driven by a duplibiotic effect, resulting from the synergistic action of the bioactive compounds.
Collapse
Affiliation(s)
- Jessica Alves Freitas
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Victor Nehmi Filho
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Aline Boveto Santamarina
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Gilson Masahiro Murata
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Clínica Médica, Laboratório de Nefrologia (LIM-29), São Paulo, SP, Brazil
| | - Lucas Augusto Moyses Franco
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
| | - Joyce Vanessa Fonseca
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM-49), São Paulo, SP, Brazil
| | - Roberta Cristina Martins
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
| | - Gabriele Alves Souza
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Gabriela Benicio
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Isabella Mirandez Sabbag
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Esther Alves de Souza
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - José Pinhata Otoch
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
- Hospital Universitário da Universidade de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
- Instituto Botânio, São Paulo, Brazil
| |
Collapse
|
6
|
Goldbaum AA, Bowers LW, Cox AD, Gillig M, Clapp Organski A, Cross TWL. The Role of Diet and the Gut Microbiota in the Obesity-Colorectal Cancer Link. Nutr Cancer 2025:1-14. [PMID: 40108862 DOI: 10.1080/01635581.2025.2476779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Obesity is positively associated with colorectal cancer (CRC) risk. Diet not only contributes to obesity, but also strongly influences the gut microbiota, a factor that is thought to independently affect CRC. To isolate the role of obesity-associated gut microbiota in CRC and to assess the impact of diet composition on this relationship, we transplanted the gut microbiota from donor mice that developed obesity or remained lean on a high-fat diet (HFD), Western diet (WD), or low-fat diet (LFD) into antibiotic-treated recipient mice that subsequently received azoxymethane to induce CRC. We hypothesized that the obesogenic diets of the donor mice, rather than their obesity status, would be a stronger driver of gut microbiota-mediated CRC development. Interestingly, while evidence supporting our hypothesis was observed, differential effects on CRC outcomes based on the type of obesogenic diets were found, such that HFD-associated gut microbiota promotes tumor incidence whereas WD-associated gut microbiota promotes tumor growth. Significantly enriched bacterial taxa present before tumor induction may be mediating these results through intestinal permeability or inflammation, such as Sutterella and Dorea in mice received HFD-associated gut microbiota, and Bacteroidetes in mice received WD-microbiota. Overall, our results demonstrated that diet drives the gut microbiota-derived impact on CRC development.
Collapse
Affiliation(s)
- Audrey A Goldbaum
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Laura W Bowers
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Abigail D Cox
- Department of Comparative Pathobiology, Purdue College of Veterinary Medicine, West Lafayette, Indiana, USA
| | - Molly Gillig
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Anna Clapp Organski
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Tzu-Wen L Cross
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
7
|
Schumacher SM, Doyle WJ, Hill K, Ochoa-Repáraz J. Gut microbiota in multiple sclerosis and animal models. FEBS J 2025; 292:1330-1356. [PMID: 38817090 PMCID: PMC11607183 DOI: 10.1111/febs.17161] [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: 10/17/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) neurodegenerative and neuroinflammatory disease marked by a host immune reaction that targets and destroys the neuronal myelin sheath. MS and correlating animal disease models show comorbidities, including intestinal barrier disruption and alterations of the commensal microbiome. It is accepted that diet plays a crucial role in shaping the microbiota composition and overall gastrointestinal (GI) tract health, suggesting an interplay between nutrition and neuroinflammation via the gut-brain axis. Unfortunately, poor host health and diet lead to microbiota modifications that could lead to significant responses in the host, including inflammation and neurobehavioral changes. Beneficial microbial metabolites are essential for host homeostasis and inflammation control. This review will highlight the importance of the gut microbiota in the context of host inflammatory responses in MS and MS animal models. Additionally, microbial community restoration and how it affects MS and GI barrier integrity will be discussed.
Collapse
Affiliation(s)
| | | | - Kristina Hill
- Department of Biological Sciences, Boise State University, Boise, ID 83725
| | | |
Collapse
|
8
|
Salvestrini V, Conti G, D'Amico F, Cristiano G, Candela M, Cavo M, Turroni S, Curti A. Gut Microbiome as a Potential Marker of Hematologic Recovery Following Induction Therapy in Acute Myeloid Leukemia Patients. Cancer Med 2025; 14:e70501. [PMID: 39865898 PMCID: PMC11770270 DOI: 10.1002/cam4.70501] [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: 08/09/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND The management of acute myeloid leukemia (AML) is hindered by treatment-related toxicities and complications, particularly cytopenia, which remains a leading cause of mortality. Given the pivotal role of the gut microbiota (GM) in hemopoiesis and immune regulation, we investigated its impact on hematologic recovery during AML induction therapy. METHODS We profiled the GM of 27 newly diagnosed adult AML patients using 16S rRNA amplicon sequencing and correlated it with key clinical parameters before and after induction therapy. RESULTS Our investigation revealed intriguing associations between the GM composition and crucial recovery indicators, including platelet, lymphocyte, and neutrophil counts, and identified early GM signatures predictive of improved hematologic recovery. Remarkably, patients demonstrating superior recovery had higher alpha diversity and enrichment in health-associated taxa belonging to the genera Faecalibacterium, Ruminococcus, Blautia, and Butyricimonas at diagnosis. CONCLUSIONS Despite certain study limitations, our findings suggest that evaluating GM features could serve as a potential marker for hematologic recovery. This preliminary work opens avenues for personalized risk assessment and interventions, possibly involving GM modulation tools, to optimize recovery in AML patients undergoing induction therapy and potentially enhancing overall outcomes in individuals with hematologic diseases.
Collapse
Affiliation(s)
- Valentina Salvestrini
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Gabriele Conti
- Human Microbiomics Unit, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Federica D'Amico
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Gianluca Cristiano
- Oncology and Haematology Research Area, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Marco Candela
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Michele Cavo
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Oncology and Haematology Research Area, Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and BiotechnologyUniversity of BolognaBolognaItaly
| | - Antonio Curti
- Istituto di Ematologia "Seràgnoli"IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| |
Collapse
|
9
|
Li F, Han Q, Cai Y, Li Y, Yang Y, Li J, Wu R, Chen R, Liu R. Si-Ni-San ameliorates cholestatic liver injury by favoring P. goldsteinii colonization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118804. [PMID: 39270883 DOI: 10.1016/j.jep.2024.118804] [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: 07/15/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Current treatment options for cholestatic liver diseases are limited, and addressing impaired intestinal barrier has emerged as a promising therapeutic approach. Si-Ni-San (SNS) is a Traditional Chinese Medicine (TCM) formula commonly utilized in the management of chronic liver diseases. Our previous studies have indicated that SNS effectively enhanced intestinal barrier function through the modulation of gut microbiota. AIM OF THE STUDY This study aims to verify the therapeutic effects of SNS on cholestatic liver injury, focusing on elucidating the underlying mechanism involving the gut-liver axis. MATERIALS AND METHODS The 16s RNA gene sequencing, non-targeted metabolomics were used to investigate the effects of SNS on the gut microbiota dysbiosis. Fecal microbiota transplantation (FMT) was conducted to identify potential beneficial probiotics underlying the therapeutic effects of SNS. RESULTS Our results demonstrated that SNS significantly ameliorated cholestatic liver injury induced by partial bile duct ligation (pBDL). Additionally, SNS effectively suppressed cholestasis-induced inflammation and barrier dysfunction in both the small intestine and colon. While SNS did not impact the intestinal FXR-FGF15-hepatic CYP7A1 axis, it notably improved gut microbiota dysbiosis and modulated the profile of microbial metabolites, including beneficial secondary bile acids and tryptophan derivatives. Furthermore, gut microbiota depletion experiments and FMT confirmed that the therapeutic benefits of SNS in cholestatic liver disease are dependent on gut microbiota modulation, particularly through the promotion of the growth of potential probiotic P. goldsteinii. Moreover, a synergistic improvement in cholestatic liver injury was observed with the co-administration of P. goldsteinii and SNS. CONCLUSION Our study underscores that SNS effectively alleviates cholestatic liver injury by addressing gut microbiota dysbiosis and enhancing intestinal barrier function, supporting its rational clinical utilization. Furthermore, we highlight P. goldsteinii as a promising probiotic candidate for the management of cholestatic liver diseases.
Collapse
Affiliation(s)
- Fanghong Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qi Han
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jianan Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiyu Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ranyun Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
10
|
Lee DB, Hwang IS. Macronutrient balance determines the human gut microbiome eubiosis: insights from in vitro gastrointestinal digestion and fermentation of eight pulse species. Front Microbiol 2025; 15:1512217. [PMID: 39949350 PMCID: PMC11823474 DOI: 10.3389/fmicb.2024.1512217] [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: 10/22/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025] Open
Abstract
The interactions between macronutrients, the human gut microbiome, and their metabolites (short-chain fatty acids) were comprehensively investigated via an in vitro digestion and fermentation model subjected to eight pulse species. 16S rRNA sequencing and taxonomic analysis of pulse digesta fermented for up to 24 h revealed an increase in the relative abundance of gut health-detrimental genera represented by Escherichia-Shigella in kidney bean, soybean, cowpea, chickpea, and black bean samples. In contrast, the relative abundance of health-positive genera, including Bacteroides, Eubacterium, and Akkermansia, was elevated in red bean, mung bean, and Heunguseul. At the same time, the proportion of the pathogenic Escherichia-Shigella decreased. Concurrently, these three species exhibited an increase in microbial diversity as evidenced by the calculation of α-diversity (Shannon index) and β-diversity (Bray-Curtis distance). Despite the lower nutrient contents in the three pulses, represented by carbohydrates, amino acids, and fatty acids, network analysis revealed that the nutrient contents in the pulse digesta possess complex positive or negative correlations with a variety of bacteria, as well as their metabolites. These correlations were more pronounced in red bean, mung bean, and Heunguseul than in the other pulses. It was postulated that the overall potential to nourish gut environments in these species was due to the balance of their nutritional components. The linear regression analysis demonstrated that there was a negative association between carbohydrate and amino acid contents and the increase in Shannon indices. Furthermore, the ratio of carbohydrates to fatty acids and amino acids to fatty acids displayed negative correlations with the diversity increase. The ratio of carbohydrates to amino acids showed a weak positive correlation. It is noteworthy that a diet comprising foods with a balanced nutritional profile supports the growth of beneficial gut microbes, thereby promoting microbial eubiosis. Consistent work on different ingredients is essential for precise insight into the interplay between food and the human microbiome in complex dietary patterns.
Collapse
Affiliation(s)
| | - In Seon Hwang
- Food and Nutrition Division, Department of Agri-food Resources, National Institute of Agricultural Sciences, Wanju, Republic of Korea
| |
Collapse
|
11
|
Shi Y, Maga EA, Mienaltowski MJ. Fecal microbiota changes associated with pathogenic and non-pathogenic diarrheas in foals. BMC Res Notes 2025; 18:34. [PMID: 39849534 PMCID: PMC11760091 DOI: 10.1186/s13104-025-07110-9] [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: 07/11/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025] Open
Abstract
OBJECTIVES Diarrhea is a common disease that could threaten the welfare of newborn foals. While there are several forms of foal diarrhea, the etiologies can be considered known pathogenic or non-pathogenic in nature. Moreover, there are likely differences in the composition of microbial populations in the gastrointestinal tracts of foals depending upon the etiology of diarrhea. Our study aims to examine the microbial population in the feces of foals with both pathogenic and non-pathogenic diarrheas to discern differences in their microbial compositions. RESULTS Foal diarrhea samples tested positive or negative for common equine neonatal diarrhea pathogens by diagnostic polymerase chain reaction (PCR), which allowed for samples to be segregated as pathogenic or non-pathogenic. Pathogenic samples tested positive for combinations of Clostridium perfringens and/or Clostridioides difficile toxins. As a result, significantly higher alpha diversity was seen in the non-pathogenic samples than in pathogenic ones. Sequencing of the V4 domains of bacterial 16 S rRNA genes demonstrated that non-pathogenic samples had more alpha diversity. Furthermore, eight microbial families and eleven genera showed significant differences in their abundances between pathogenic and non-pathogenic diarrhea samples.
Collapse
Affiliation(s)
- Yijun Shi
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Elizabeth A Maga
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Michael J Mienaltowski
- Department of Animal Science, University of California Davis, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
12
|
Jeong S, Tollison TS, Brochu H, Chou H, Huntress I, Yount KS, Zheng X, Darville T, O'Connell CM, Peng X. Cervicovaginal microbial features predict Chlamydia trachomatis spread to the upper genital tract of infected women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625070. [PMID: 39651251 PMCID: PMC11623589 DOI: 10.1101/2024.11.26.625070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Chlamydia trachomatis (CT) infection can lead to pelvic inflammatory disease, infertility and other reproductive sequelae when it ascends to the upper genital tract. Factors including chlamydial burden, co-infection with other sexually-transmitted bacterial pathogens and oral contraceptive use influence risk for upper genital tract spread. Cervicovaginal microbiome composition influences CT susceptibility and we investigated if it contributes to spread by analyzing amplicon sequence variants (ASVs) derived from the V4 region of 16S rRNA genes in vaginal samples collected from women at high risk for CT infection and for whom endometrial infection had been determined. RESULTS Participants were classified as CT negative (CT-, n=77), CT positive at the cervix (Endo-, n=77), or CT positive at both cervix and endometrium (Endo+, n=66). Although we were unable to identify many significant differences between CT infected and uninfected women, differences in abundance of ASVs representing Lactobacillus iners and L. crispatus subspecies but not dominant lactobacilli were detected. Twelve informative ASVs predicted endometrial chlamydial infection (AUC=0.74), with CT ASV abundance emerging as a key predictor. We also observed a positive correlation between levels of cervically secreted cytokines previously associated with CT ascension and abundance of the informative ASVs. CONCLUSION Our findings suggest that vaginal microbial community members may influence chlamydial spread directly by nutrient limitation and/or disrupting endocervical epithelial integrity and indirectly by modulating pro-inflammatory signaling and/or homeostasis of adaptive immunity. Further investigation of these predictive microbial factors may lead to cervicovaginal microbiome biomarkers useful for identifying women at increased risk for disease.
Collapse
|
13
|
Ying J, Zhang MW, Wei KC, Wong SH, Subramaniam M. Influential articles in autism and gut microbiota: bibliometric profile and research trends. Front Microbiol 2025; 15:1401597. [PMID: 39850141 PMCID: PMC11755156 DOI: 10.3389/fmicb.2024.1401597] [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: 04/16/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
Objective Autism spectrum disorder (ASD) is a common neurodevelopmental disorder. Increasing evidence suggests that it is potentially related to gut microbiota, but no prior bibliometric analysis has been performed to explore the most influential works in the relationships between ASD and gut microbiota. In this study, we conducted an in-depth analysis of the most-cited articles in this field, aiming to provide insights to the existing body of research and guide future directions. Methods A search strategy was constructed and conducted in the Web of Science database to identify the 100 most-cited papers in ASD and gut microbiota. The Biblioshiny package in R was used to analyze and visualize the relevant information, including citation counts, country distributions, authors, journals, and thematic analysis. Correlation and comparison analyses were performed using SPSS software. Results The top 100 influential manuscripts were published between 2000 and 2021, with a total citation of 40,662. The average number of citations annually increased over the years and was significantly correlated to the year of publication (r = 0.481, p < 0.01, Spearman's rho test). The United States was involved in the highest number of publications (n = 42). The number of publications in the journal was not significantly related to the journal's latest impact factor (r = 0.016, p > 0.05, Spearman's rho test). Co-occurrence network and thematic analysis identified several important areas, such as microbial metabolites of short-chain fatty acids and overlaps with irritable bowel syndrome. Conclusion This bibliometric analysis provides the key information of the most influential studies in the area of ASD and gut microbiota, and suggests the hot topics and future directions. The findings of this study can serve as a valuable reference for researchers and policymakers, guiding the development and implementation of the scientific research strategies in this area.
Collapse
Affiliation(s)
- Jiangbo Ying
- Department of Developmental Psychiatry, Institute of Mental Health, Singapore, Singapore
| | | | - Ker-Chiah Wei
- Department of Developmental Psychiatry, Institute of Mental Health, Singapore, Singapore
| | - Sunny H. Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore, Singapore
| | | |
Collapse
|
14
|
Vogel CL, Geary EL, Oba PM, Mioto JC, Rudolph BC, Rens L, Swanson KS. Effects of corn protein inclusion on apparent total tract macronutrient digestibility, palatability, and fecal characteristics, microbiota, and metabolites of healthy adult dogs. J Anim Sci 2025; 103:skaf122. [PMID: 40243089 PMCID: PMC12065405 DOI: 10.1093/jas/skaf122] [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: 01/25/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025] Open
Abstract
Corn protein (CP), a co-product of the corn ethanol industry, is a sustainable protein source used in pet foods. The objectives of this study were to determine the palatability and apparent total tract digestibility (ATTD) of diets containing CP and to test the effects of CP-containing diets on the serum metabolites, hematology, and fecal characteristics, metabolites, and microbiota of healthy adult dogs. Ten female adult beagles (mean age: 6.12 ± 1.39 yr; mean body weight: 9.33 ± 1.04 kg) were used in a replicated 5 × 5 Latin square design (n = 10/treatment). All dietary treatments were based on brewers rice, low-ash chicken byproduct meal, and chicken fat, and contained variable amounts of corn-based proteins: 0% (control), 15.3% corn gluten meal (CGM), 10.2% CGM + 5% CP (Low), 5.1% CGM + 10% CP (Medium), and 15% CP (High). The experiment was composed of five 28-d periods, with each consisting of a 22-d diet consumption period, a 5-d fecal collection period, and 1 d for blood collection. Data were analyzed statistically by Mixed Models using SAS 9.4, with P < 0.05 accepted as being statistically significant. Two 2-d palatability studies (n = 20 dogs) were also conducted to compare the High diet vs. control diet and High diet vs. CGM diet. Dogs were shown to prefer (P < 0.05) the High diet over the control diet by a ratio of 1.8:1, but no preference was observed between the High and CGM diets. In the digestibility study, the ATTD of dry matter, organic matter, and energy increased (P < 0.001) linearly with CP inclusion. The ATTD of fat was greater (P < 0.001) for the control diet than for the CGM, Low, and High diets. Fecal scores were lower (P = 0.05; firmer) and fecal dry matter percentage was higher (P < 0.0001) in dogs fed CGM than those fed CP. Fecal phenol and indole, short-chain fatty acid, and branched-chain fatty acid concentrations were greater (P < 0.05) in dogs fed CP than dogs fed CGM and control. Fecal bacterial diversity was not altered by diet, but the relative abundance of approximately 10 bacterial genera was altered by diet. In summary, our data demonstrate that the inclusion of CP in dog foods resulted in high diet palatability and macronutrient digestibility and altered microbial composition and activity.
Collapse
Affiliation(s)
- Christina L Vogel
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Elizabeth L Geary
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Patrícia M Oba
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Julio C Mioto
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | | | - Louis Rens
- Green Plains, Inc., Omaha, NE 68106, USA
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Veterinary Clinical Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
15
|
Misera A, Kaczmarczyk M, Łoniewski I, Liśkiewicz P, Podsiadło K, Misiak B, Skonieczna-Żydecka K, Samochowiec J. Comparative analysis of gut microbiota in major depressive disorder and schizophrenia during hospitalisation - the case-control, post hoc study. Psychoneuroendocrinology 2025; 171:107208. [PMID: 39426041 DOI: 10.1016/j.psyneuen.2024.107208] [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: 05/06/2024] [Revised: 08/14/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024]
Abstract
The aim of this study was to investigate the relationship between gut microbiota and major depressive disorder (MDD) and schizophrenia (SCZ) by comparing 36 inpatients with these conditions to 29 healthy controls (HC) matched for age, sex, and body mass index (BMI). Individuals with SCZ exhibited greater microbiota richness compared to HC (FDR P(Q)=0.028). Taxonomically, while no significant differences were observed between the microbiota of MDD and SCZ patients in a head-to-head comparison, both patient groups differed significantly when compared to HC. Interestingly, besides common patterns (such as a higher abundance of Erysipelotrichaceae UCG-003 and Streptococcus, and a lower abundance of Lachnospiraceae ND3007 group), unique patterns were exhibited only in MDD (with a higher abundance of Anaerostipes, Q=0.004) or SCZ (with a higher abundance of Sutterella, Q=0.001, and a lower abundance of Clostridium sensu stricto 1, Q=0.002). The Random Forest algorithm identified Ruminococcus torques group, Lachnospiraceae UCG-001, and Erysipelotrichaceae UCG-003 as highly discriminative features for both SCZ and MDD, while Suturella and Holdemania were unique features for SZC, and Lachnospiraceae genus CAG-56 and Anaerostipes for MDD. Additionally, between 50 % and 60 % of the differentially abundant taxa were found among the top 10 influential features in the RF models. In conclusion, while no significant differences were found between the microbiota of MDD and SCZ patients, distinct microbial patterns were found in each group when compared to HC. The study did not confirm universal microbial biomarkers reported in other studies but showed that the observed differences concern the bacteria associated with inflammation, the production of short chain fatty acids (SCFA), and the synthesis of metabolites linked to mental health (lactic acid, gamma-aminobutyric acid - GABA). The application of machine learning holds promise for further understanding the complex relationship between microbiota and these psychiatric disorders. The observed results should be treated with caution due to the limitations of this study (mainly sample size), therefore further researches under standardized environmental conditions with consistent analytical and bioinformatics approaches are warranted.
Collapse
Affiliation(s)
- Agata Misera
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Sanprobi sp. z o. o. sp. k, Szczecin, Poland; Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Igor Łoniewski
- Sanprobi sp. z o. o. sp. k, Szczecin, Poland; Department of Biochemical Science, Pomeranian Medical University in Szczecin, Szczecin, Poland.
| | - Paweł Liśkiewicz
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Błażej Misiak
- Department of Psychiatry, Wrocław Medical University, Wrocław, Poland
| | | | - Jerzy Samochowiec
- Department of Psychiatry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
16
|
Cheng SH, Yang YC, Chen CP, Hsieh HT, Lin YC, Cheng CY, Liao KS, Chu FY, Liu YR. Oncogenic human papillomavirus and anal microbiota in men who have sex with men and are living with HIV in Northern Taiwan. PLoS One 2024; 19:e0304045. [PMID: 39739827 DOI: 10.1371/journal.pone.0304045] [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: 05/06/2024] [Accepted: 10/07/2024] [Indexed: 01/02/2025] Open
Abstract
Few studies have demonstrated the interplay between human immunodeficiency virus (HIV), anal human papillomavirus (HPV), and anal microbiota, especially in persons living with HIV who are men who have sex with men. We, therefore, explored these interrelationships in a cohort of persons living with HIV, mainly comprising men who have sex with men. HPV genotyping using a commercial genotyping kit and ThinPrep cytology interpreted by Bethesda systems was performed on samples from 291 patients. Samples were characterized by high-throughput sequencing of dual-index barcoded 16s rRNA (V3-4). Bacterial diversity was diminished in individuals living with HIV with CD4+ T cells <500 cells/μL and anal cytology yielding atypical squamous cells of undetermined significance or higher grades (ASCUS+) with detectable HPV 16/18 compared with those with CD4+ T cells ≥500 cells/μL with ASCUS+ and HPV 16/18 and those with normal anal cytology or inflammation without HPV 16/18. Enterobacteriaceae, Ruminococcus, and Bacilli were significantly abundant in persons living with HIV with CD4+ T cells <500 cells/μL with ASCUS+ and HPV 16/18. Bacterial diversity, composition, and homogeneity of dispersion were different in individuals living with HIV with low CD4+ T cells with ASCUS+ and HPV 16/18, and understanding the interaction among immunocompromised hosts, oncogenic HPVs, and microbiota is essential, and the contribution of these factors to anal precancerous lesions needs more in-depth exploration.
Collapse
Affiliation(s)
- Shu-Hsing Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Pin Chen
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hui-Ting Hsieh
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Yi-Chun Lin
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Sheng Liao
- Department of Pathology, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
| | - Fang-Yeh Chu
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
17
|
Huang Z, Wang L, Tong J, Zhao Y, Ling H, Zhou Y, Tan Y, Xiong X, Qiu Y, Bi Y, Pan Z, Yang R. Alterations in Gut Microbiota Correlate With Hematological Injuries Induced by Radiation in Beagles. Int J Microbiol 2024; 2024:3096783. [PMID: 39659556 PMCID: PMC11631345 DOI: 10.1155/ijm/3096783] [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: 03/05/2024] [Revised: 09/04/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Dynamics of gut microbiota and their associations with the corresponding hematological injuries postradiation remain to be elucidated. Using single whole-body exposure to 60Co-γ ray radiation at the sublethal dose of 2.5 Gy, we developed a beagle model of acute radiation syndrome (ARS) and then monitored the longitudinal changes of gut microbiome and hematology for 45 days. We found that the absolute counts of circulating lymphocytes, neutrophils, and platelets were sharply declined postradiation, accompanied by a largely shifted composition of gut microbiome that manifested as a significantly increased ratio of Firmicutes to Bacteroidetes. In irradiated beagles, alterations in hematological parameters reached a nadir on day 14, sustaining for 1 week, which were gradually returned to the normal levels thereafter. However, no structural recovery of gut microbiota was observed throughout the study. Fecal metagenomics revealed that irradiation increased the relative abundances of genus Streptococcus, species Lactobacillus animalis and Lactobacillus murinus, but decreased those of genera Prevotella and Bacteroides. Metagenomic functions prediction demonstrated that 26 altered KEGG pathways were significantly enriched on Day 14 and 35 postradiation. Furthermore, a total of 43 bacterial species were found to correlate well with hematological parameters by Spearman's analysis. Our results provide an insight into the longitudinal changes in intestinal microbiota at different clinical stages during ARS in canine. Several key microbes those tightly associated with the hematological alterations may serve as biomarkers to discriminate the different phases of host with ARS.
Collapse
Affiliation(s)
- Zongyu Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 210009, Jiangsu, China
| | - Likun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Jianghui Tong
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Yong Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Hui Ling
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
- Department of Microbiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230000, Anhui, China
| | - Yazhou Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Yafang Tan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Xiaohui Xiong
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 210009, Jiangsu, China
| | - Yefeng Qiu
- Laboratory Animal Center, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| | - Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
- Department of Research and Development, Grand Life Sciences Group Ltd., China Grand Enterprises Inc., Chaoyang, Beijing 100101, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Fengtai, Beijing 100071, China
| |
Collapse
|
18
|
Yang KC, Tien WY, Cheng MF. Gut microbiota compositions in the carriers and noncarriers of third-generation cephalosporin-resistant Escherichia coli: A study among children in southern Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024; 57:896-905. [PMID: 39261124 DOI: 10.1016/j.jmii.2024.08.010] [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: 04/16/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Antimicrobial resistance, particularly in third-generation cephalosporin-resistant (3GC-R) Escherichia coli (E. coli), poses major global health challenges and has various clinical implications. Researchers have explored the relationship between extended-spectrum β-lactamase-producing E. coli and gut microbiota composition, which influence host health and disease susceptibility, in adults. In this study, we analyzed gut microbiota composition in Taiwanese children by the colonization status of 3GC-R E. coli. METHODS This cross-sectional study included children (age, 0-6 years) from Kaohsiung, Taiwan. Fecal samples were subjected to microbiological and gut microbiome (full-length 16S rRNA sequencing) analyses. The antimicrobial susceptibility of E. coli colonies isolated from the samples was tested. Furthermore, gut microbiota compositions and diversity indices were compared between 3GC-R E. coli carriers and noncarriers. RESULTS Approximately 46% of all children aged <6 years carried 3GC-R E. coli. The abundances of Drancourtella, Romboutsia, and Desulfovibrio (genus level) were higher in carriers than in noncarriers. By contrast, the abundances of Odoribacteraceae (family level) and Sutterella (genus level) were higher in noncarriers than in carriers. No significant between-group difference was observed in alpha diversity. However, a significant between-group difference was noted in beta diversity (unweighted UniFrac analysis). CONCLUSION This is the first study that investigated differences in the gut microbiota between healthy 3GC-R E. coli carriers and noncarriers in children, suggesting potential mechanisms involving altered utilization of short-chain fatty acids and elevated succinate levels contributing to increased colonization of 3GC-R E. coli. The other taxa identified in this study may contribute to colonization resistance in the pediatric population.
Collapse
Affiliation(s)
- Keng-Chin Yang
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wan-Yu Tien
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ming-Fang Cheng
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Kaohsiung Veterans General Hospital Tainan Branch, Tainan, Taiwan.
| |
Collapse
|
19
|
McCoubrey LE, Shen C, Mwasambu S, Favaron A, Sangfuang N, Thomaidou S, Orlu M, Globisch D, Basit AW. Characterising and preventing the gut microbiota's inactivation of trifluridine, a colorectal cancer drug. Eur J Pharm Sci 2024; 203:106922. [PMID: 39368784 DOI: 10.1016/j.ejps.2024.106922] [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: 06/17/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The gut microbiome can metabolise hundreds of drugs, potentially affecting their bioavailability and pharmacological effect. As most gut bacteria reside in the colon, drugs that reach the colon in significant proportions may be most impacted by microbiome metabolism. In this study the anti-colorectal cancer drug trifluridine was used as a model drug for characterising metabolism by the colonic microbiota, identifying correlations between bacterial species and individuals' rates of microbiome drug inactivation, and developing strategies to prevent drug inactivation following targeted colonic delivery. High performance liquid chromatography and ultra-high performance liquid chromatography coupled with high resolution tandem mass spectrometry demonstrated trifluridine's variable and multi-route metabolism by the faecal microbiota sourced from six healthy humans. Here, four drug metabolites were linked to the microbiome for the first time. Metagenomic sequencing of the human microbiota samples revealed their composition, which facilitated prediction of individual donors' microbial trifluridine inactivation. Notably, the abundance of Clostridium perfringens strongly correlated with the extent of trifluridine inactivation by microbiota samples after 2 hours (R2 = 0.8966). Finally, several strategies were trialled for the prevention of microbial trifluridine metabolism. It was shown that uridine, a safe and well-tolerated molecule, significantly reduced the microbiota's metabolism of trifluridine by acting as a competitive enzyme inhibitor. Further, uridine was found to provide prebiotic effects. The findings in this study greatly expand knowledge on trifluridine's interactions with the gut microbiome and provide valuable insights for investigating the microbiome metabolism of other drugs. The results demonstrate how protection strategies could enhance the colonic stability of microbiome-sensitive drugs.
Collapse
Affiliation(s)
- Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Chenghao Shen
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Sydney Mwasambu
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Alessia Favaron
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Nannapat Sangfuang
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Stavrina Thomaidou
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Mine Orlu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Daniel Globisch
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom.
| |
Collapse
|
20
|
Shi L, Gao P, Zhang Y, Liu Q, Hu R, Zhao Z, Hu Y, Xu X, Shen Y, Liu J, Long J. 2-(3,4-Dihydroxyphenyl)ethyl 3-hydroxybutanoate Ameliorates Cognitive Dysfunction and Inflammation Via Modulating Gut Microbiota in Aged Senescence-Accelerated Mouse Prone8 Mice. J Gerontol A Biol Sci Med Sci 2024; 79:glae220. [PMID: 39215682 DOI: 10.1093/gerona/glae220] [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: 02/19/2024] [Indexed: 09/04/2024] Open
Abstract
Numerous studies have indicated a close association between gut microbiota dysbiosis, inflammation, and cognitive impairment, highlighting their crucial role in the aging process. 2-(3,4-Dihydroxyphenyl)ethyl 3-hydroxybutanoate (HTHB), a novel derivative of hydroxytyrosol, known for its metabolic and anti-inflammatory properties, was investigated for its effects on memory, inflammation, and gut microbiota in senescence-accelerated mouse prone 8 mice. The study employed behavioral testing, biochemical detection, and 16S RNA analysis. Results revealed that HTHB mitigated memory decline and lymphocyte aberrance, reduced inflammation in the brain cortex, intestine and peripheral system, and modulated gut microbiota dysbiosis. Interestingly, the cognitive function and serum inflammation of mice significantly correlated with differences in gut microbiota in senescence-accelerated mouse prone 8 mice. Furthermore, HTHB treatment exhibited an enhancement of gut barrier integrity in colon tissue in SAMP8 mice. In vitro experiments using HCT116 and DLD1 cells further evidenced that HTHB rescued the tight junction protein levels impaired by lipopolysaccharide. These findings demonstrate that HTHB effectively ameliorates cognitive dysfunction in aged mice, by modulating gut microbiota, suppressing inflammation, and promoting intestinal barrier integrity. This highlights the potential of HTHB as a therapeutic agent for age-related cognitive loss.
Collapse
Affiliation(s)
- Le Shi
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peipei Gao
- Department of Health Education and Management, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, China
| | - Yue Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Quanyu Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ranrui Hu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhuang Zhao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yachong Hu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaohong Xu
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan, China
| | - Yehua Shen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi, China
| | - Jiankang Liu
- Department of Dermatology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
21
|
Aluthge N, Adams S, Davila CA, Gocchi Carrasco NR, Chiou KS, Abadie R, Bennett SJ, Dombrowski K, Major AM, Valentín-Acevedo A, West JT, Wood C, Fernando SC. Gut microbiota profiling in injection drug users with and without HIV-1 infection in Puerto Rico. Front Microbiol 2024; 15:1470037. [PMID: 39697649 PMCID: PMC11652967 DOI: 10.3389/fmicb.2024.1470037] [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: 07/24/2024] [Accepted: 10/23/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction The full extent of interactions between human immunodeficiency virus (HIV) infection, injection drug use, and the human microbiome is unclear. In this study, we examined the microbiomes of HIV-positive and HIV-negative individuals, both drug-injecting and non-injecting, to identify bacterial community changes in response to HIV and drug use. We utilized a well-established cohort of people who inject drugs in Puerto Rico, a region with historically high levels of injection drug use and an HIV incidence rate disproportionately associated with drug use. Methods Using amplicon-based 16S rDNA sequencing, we identified amplicon sequence variants (ASVs) that demonstrated significant variations in the composition of microbial communities based on HIV status and drug use. Results and discussion Our findings indicate that the HIV-positive group exhibited a higher abundance of ASVs belonging to the genera Prevotella, Alloprevotella, Sutterella, Megasphaera, Fusobacterium, and Mitsuokella. However, Bifidobacteria and Lactobacillus ASVs were more abundant in injectors than in non-injectors. We examined the effect of drug use on the gut microbiome in both HIV-infected and non-infected patients, and found that multiple drug use significantly affected the microbial community composition. Analysis of differential of bacterial taxa revealed an enrichment of Bifidobacterium spp., Faecalibacterium spp., and Lactobacillus spp. in the multiple drug-injecting group. However, in the non-injecting group, Parabacteroides spp., Prevotella spp., Paraprevotella spp., Sutterella spp., and Lachnoclostridium spp. The presence of multiple drug-injecting groups was observed to be more prevalent. Our findings provide detailed insight into ASV-level changes in the microbiome in response to HIV and drug use, suggesting that the effect of HIV status and drug injection may have different effects on microbiome composition and in modulating gut bacterial populations.
Collapse
Affiliation(s)
- Nirosh Aluthge
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Seidu Adams
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Carmen A. Davila
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - Kathy S. Chiou
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Roberto Abadie
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, United States
| | - Sydney J. Bennett
- Department of Biological Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | | | - Angel M. Major
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamon, Puerto Rico
| | - Aníbal Valentín-Acevedo
- Department of Microbiology and Immunology, Universidad Central del Caribe, Bayamon, Puerto Rico
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, United States
| | - Charles Wood
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA, United States
| | - Samodha C. Fernando
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
22
|
Bai B, Luo L, Yao F, Sun Q, Chen X, Zheng W, Jiang L, Wang X, Su G. The causal relationship between the human gut microbiota and pyogenic arthritis: a Mendelian randomization study. Front Cell Infect Microbiol 2024; 14:1452480. [PMID: 39660282 PMCID: PMC11629706 DOI: 10.3389/fcimb.2024.1452480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Background Recent studies have indicated the role of the gut microbiota in the progression of osteoarticular diseases, however, the causal relationship between the gut microbiota and pyogenic arthritis remains unclear. There is also a lack of theoretical basis for the application of the gut microbiota in the treatment of pyogenic arthritis. Methods In our study, we utilized the largest genome-wide association study (GWAS) data from the MiBioGen Consortium involving 13,400 participants and extracted summary statistical data of the microbiota metabolic pathways of 7,738 participants of European descent from the Dutch Microbiome Project (DMP) The data of pyogenic arthritis were derived from the FinnGen R10 database, including 1,086 patients and 147,221 controls. We employed the two-sample Mendelian randomization approach to investigate the causal association between the gut microbiota and pyogenic arthritis. Our methods comprised inverse variance weighting, Mendelian Randomization Egger regression, weighted median, and weighted modal methods. Subsequently, polygenic and heterogeneity analyses were conducted. Results At the class level, β-proteobacteria is positively correlated with the risk of pyogenic arthritis. At the order level, Burkholderia is positively associated with the disease. At the genus level, the unclassified genus of Sutterellaceae is positively correlated with the disease, while the unnamed genus of Lachnospiraceae, Rothia, and the unnamed genus of Erysipelotrichaceae are negatively correlated with the disease. In addition, Faecalibacterium and Finegoldia are also negatively correlated with the disease. Sensitivity analysis did not show any abnormal evidence. Conclusion This study indicates that β-proteobacteria, Burkholderiales, and the unclassified genus of Sutterellaceae are associated with an increased risk of the disease, while the unnamed genus of Lachnospiraceae, Rothia, the unnamed genus of Erysipelotrichaceae, Faecalibacterium, and Finegoldia are related to a reduced risk. Future studies are needed to elucidate the specific mechanisms by which these specific bacterial groups affect pyogenic arthritis.
Collapse
Affiliation(s)
- Boliang Bai
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Longfei Luo
- Department of Bioinformatics, Center for Systems Biology, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Feng Yao
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Qian Sun
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Xingguang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wen Zheng
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Lang Jiang
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Xiaodong Wang
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Guanghao Su
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Petrilla A, Nemeth P, Fauszt P, Szilagyi-Racz A, Mikolas M, Szilagyi-Tolnai E, David P, Stagel A, Gal F, Gal K, Sohajda R, Pham T, Stundl L, Biro S, Remenyik J, Paholcsek M. Comparative analysis of the postadmission and antemortem oropharyngeal and rectal swab microbiota of ICU patients. Sci Rep 2024; 14:27179. [PMID: 39516251 PMCID: PMC11549221 DOI: 10.1038/s41598-024-78102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Shotgun metabarcoding was conducted to examine the microbiota in a total of 48 samples from 12 critically ill patients, analyzing samples from both the oropharynx and rectum. We aimed to compare their postadmission microbiota, characterized as moderately dysbiotic, with the severely dysbiotic antemortem microbiota associated with patients' deaths. We found that, compared with postadmission samples, patient antemortem swab samples presented moderate but not significantly decreased diversity indices. The antemortem oropharyngeal samples presented an increase in biofilm-forming bacteria, including Streptococcus oralis, methicillin-resistant Staphylococcus aureus (MRSA), and Enterococcus faecalis. Although the septic shock rate was 67%, no significant differences were detected in the potential pathogen ratios when the microbiota was analyzed. A notable strain-sharing rate between the oropharynx and intestine was noted. By comparing postadmission and antemortem samples, microbial biomarkers of severe dysbiosis were pinpointed through the analysis of differentially abundant and uniquely emerging species in both oropharyngeal and rectal swabs. Demonstrating strong interconnectivity along the oral-intestinal axis, these biomarkers could serve as indicators of the progression of dysbiosis. Furthermore, the microbial networks of the oropharyngeal microbiota in deceased patients presented the lowest modularity, suggesting a vulnerable community structure. Our data also highlight the critical importance of introducing treatments aimed at enhancing the resilience of the oral cavity microbiome, thereby contributing to better patient outcomes.
Collapse
Affiliation(s)
- Annamaria Petrilla
- Department of Anaesthesiology and Intensive Care, Vas County Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Peter Nemeth
- Department of Anaesthesiology and Intensive Care, Vas County Markusovszky Teaching Hospital, Szombathely, Hungary
| | - Peter Fauszt
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Anna Szilagyi-Racz
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Maja Mikolas
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Emese Szilagyi-Tolnai
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Peter David
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Aniko Stagel
- Hungarian National Blood Transfusion Service Nucleic Acid Testing Laboratory, Budapest, Hungary
| | - Ferenc Gal
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Kristof Gal
- Department of Oncoradiology, University of Debrecen Clinical Centre, Debrecen, Hungary
| | - Reka Sohajda
- Hungarian National Blood Transfusion Service Nucleic Acid Testing Laboratory, Budapest, Hungary
| | - Trinh Pham
- Turku Bioscience Centre, University of Turku and Abo Akademi University, 20520, Turku, Finland
| | - Laszlo Stundl
- Faculty of Agricultural and Food Sciences and Environmental Management, University of Debrecen, Debrecen, Hungary
| | - Sandor Biro
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Remenyik
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary
| | - Melinda Paholcsek
- Faculty of Agricultural and Food Sciences and Environmental Management, Complex Systems and Microbiome-innovations Centre, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
24
|
Wang L, Zheng W, Men Q, Ren X, Song S, Ai C. Curcumin-loaded polysaccharide microparticles alleviated DSS-induced ulcerative colitis by improving intestinal microecology and regulating MAPK/NF-κB/Nrf2/NLRP3 pathways. Int J Biol Macromol 2024; 281:136687. [PMID: 39427805 DOI: 10.1016/j.ijbiomac.2024.136687] [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: 05/13/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Curcumin (Cur) exerts many benefits on the host, but its application is limited by its poor bioavailability. In this study, composite polysaccharide microparticles loading Cur (Cur-CPM) was prepared by food-grade materials and gel technology. Its properties were analyzed via the in vitro and in vivo models, and then its benefit on gut health was assessed in DSS-treated mice. Compared to free Cur, CPM extended the residence time and absorption efficiency of Cur in the intestine, effectively ameliorating the symptoms of colitis. Cur-CPM alleviated colonic inflammation by inhibiting the activation of the MAPK and NF-κB pathways and suppressing NLRP3 inflammasome activity, affecting the expression of inflammation-related cytokines and mediators. In addition, Cur-CPM regulated the levels of antioxidants and oxidants in the colon tissues via Nrf2 activation, alleviating oxidative stress. Cur-CPM protected gut barrier function by maintaining the integrity of colonic mucosal layer and tight junction. The underlying mechanism can be attributed not only to the anti-inflammatory and antioxidant activities of Cur but also to modulation of Cur and CPM on the gut microbiota and metabolites. It suggests that Cur-CPM holds the potential to be developed as a functional component to enhance gut health.
Collapse
Affiliation(s)
- Lu Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong 037009, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
25
|
Olbjørn C, Hagen M, Moen AEF, Havdal LB, Sommen SL, Berven LL, Thiis-Evensen E, Stiansen-Sonerud T, Selvakumar J, Wyller VBB. Longitudinal Fecal Microbiota Profiles in A Cohort of Non-Hospitalized Adolescents and Young Adults with COVID-19: Associations with SARS-CoV-2 Status and Long-Term Fatigue. Pathogens 2024; 13:953. [PMID: 39599506 PMCID: PMC11597601 DOI: 10.3390/pathogens13110953] [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: 09/21/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Adolescents most often experience mild acute COVID-19, but may still face fatigue and persistent symptoms such as post-COVID-19 condition (PCC) and post-infective fatigue syndrome (PIFS). We explored the fecal microbiota of SARS-CoV-2 positive and negative non-hospitalized adolescents and young adults (12-25 years of age) in the "Long-Term Effects of COVID-19 in Adolescents" (LoTECA) project, a longitudinal observational cohort study. With a targeted qPCR approach, the quantities of 100 fecal bacterial taxa were measured at baseline (early convalescent stage) in 145 SARS-CoV-2-positive and 32 SARS-CoV-2 negative participants and after six months in 107 of the SARS-CoV-2-positive and 19 of the SARS-CoV-2 negative participants. Results: Faecalibacterium prausnitzii M21.2 and Gemmiger formicilis (both p < 0.001) were enriched in the SARS-CoV-2-positive participants compared to negative controls at baseline. In SARS-CoV-2-positive participants, lower baseline abundance of Faecalibacterium prausnitzii M21/2 (p = 0.013) and higher abundance of Clostridium spiroforme (p = 0.006), Sutterella wadsworthensis (p < 0.001), and Streptococcus thermophilus (p = 0.039) were associated with six-month fatigue. Sutterella wadsworthensis and Streptococcus thermophilus enrichment was additionally associated with PCC in the SARS-CoV-2-positive group (p < 0.001 and 0.042 respectively). Conclusions: Adolescents and young adults with mild acute COVID-19 infection had increased fecal abundance of the beneficial Faecalibacterium prausnitzii M21/2 and Gemmiger formicilis compared to SARS-CoV-2 negative controls in the early convalescent stage. Additionally, the abundance of both known (Faecalibacterium prausnitzii, Streptococcus thermophilus) and new (Clostridium spiroforme, Sutterella wadsworthensis) bacteria were associated with persistent symptoms such as fatigue in the COVID-19 infected group, warranting further exploration of the role of these bacteria in COVID-19 disease and PCC pathophysiology.
Collapse
Affiliation(s)
- Christine Olbjørn
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
| | - Milada Hagen
- Department of Nursing and Health Promotion, Oslo Metropolitan University, 0130 Oslo, Norway;
| | | | - Lise Beier Havdal
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
| | - Silke Lauren Sommen
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Lise Lund Berven
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
- Department of Microbiology and Infection Control, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Espen Thiis-Evensen
- Department of Gastroenterology, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway;
| | - Tonje Stiansen-Sonerud
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
- Department of Clinical Molecular Biology (EpiGen), University of Oslo, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Joel Selvakumar
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Vegard Bruun Bratholm Wyller
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, 1478 Lørenskog, Norway; (L.B.H.); (S.L.S.); (L.L.B.); (T.S.-S.); (J.S.); (V.B.B.W.)
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
26
|
Lee JJ, Kim KM, Kim HJ, Sohn J, Song JW, Koo HY, Lee S. The Consumption of Lacticaseibacillus rhamnosus HDB1258 Changes Human Gut Microbiota and Induces Immune Enhancement Through NK Cell Activation. Microorganisms 2024; 12:2109. [PMID: 39458418 PMCID: PMC11510592 DOI: 10.3390/microorganisms12102109] [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: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota can play an important role in enhancing the host's complex immune system. In this regard, many studies indicate that probiotics consumption has a beneficial impact on alterations in the composition of the gut microbiota. Our previous study demonstrated that the oral administration of Lacticaseibacillus rhamnosus HDB1258 (HDB1258) enhances immune cell activity and alters the composition of gut microbiota in C57BL/6 mice, thereby showing its potential as a novel immunostimulatory ingredient. Therefore, this clinical trial assessed the effects of HDB1258 on human natural killer (NK) cell activity and changes in gut microbiota. It also investigated the correlation between gut microbiota and NK cell activity following HDB1258 supplementation. Participants (n = 71) were randomized into placebo and HDB1258 groups, and NK cell activity and gut microbiota were investigated at baseline (week 0) and endline (week 8). The present study showed that HDB1258 significantly increased NK cell activity and resulted in positive regulatory effects on the gut microbial balance in subjects compared to the placebo group. HDB1258 affected the gut microbial balance by inducing the growth of beneficial bacteria such as Lactococcus and Sutterella. Especially, the changes in Escherichia-Shigella composition were negatively correlated with the changes in NK cell activity after HDB1258 consumption. There was also a positive correlation between the NK cell activity in the HDB1258 group and the composition of Prevotella 9 and Adlercreutzia. These findings suggest that HDB1258 may improve the host's intestinal environment by regulating gut bacteria related to immune response and promote NK cell activation. This study was registered at clinical research information service (CRIS: KCT0008204).
Collapse
Affiliation(s)
- Jin-Joo Lee
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| | - Kyung-Min Kim
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| | - Hyeon-Jeong Kim
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| | - Johann Sohn
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| | - Ji-Won Song
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| | - Hye-Yeon Koo
- Department of Family Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Family Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seunghun Lee
- Biohealthcare R&D Center, Hyundai Bioland Co., Ltd., Manhae-ro, Danwon-gu, Ansan 15407, Republic of Korea; (J.-J.L.); (H.-J.K.); (J.S.); (J.-W.S.)
| |
Collapse
|
27
|
Leibovitzh H, Sarbagili Shabat C, Hirsch A, Zittan E, Mentella MC, Petito V, Cohen NA, Ron Y, Fliss Isakov N, Pfeffer J, Yaakov M, Fanali C, Turchini L, Masucci L, Quaranta G, Kolonimos N, Godneva A, Weinberger A, Scaldaferri F, Maharshak N. Faecal Transplantation for Ulcerative Colitis From Diet Conditioned Donors Followed by Dietary Intervention Results in Favourable Gut Microbial Profile Compared to Faecal Transplantation Alone. J Crohns Colitis 2024; 18:1606-1614. [PMID: 38720628 DOI: 10.1093/ecco-jcc/jjae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/17/2024] [Accepted: 05/08/2024] [Indexed: 10/17/2024]
Abstract
BACKGROUND AND AIMS Several faecal microbial transplantation [FMT] approaches for ulcerative colitis [UC] have been investigated with conflicting results. We have recently published the clinical outcomes from the CRAFT UC Trial using FMT with the UC Exclusion Diet [UCED], compared with FMT alone. Here we aimed to compare the two FMT strategies in terms of microbial profile and function. METHODS Subjects recruited to the CRAFT UC study with available pre- and post-intervention faecal samples were included. Donors received diet conditioning for 14 days based on the UCED principles. Group 1 received single FMT by colonoscopy [Day 1] and enemas [Days 2 and 14] without donors' dietary conditioning [N = 11]. Group 2 received FMT but with donors' dietary pre-conditioning and UCED for the patients [N = 10]. Faecal samples were assessed by DNA shotgun metagenomic sequencing. RESULTS Following diet conditioning, donors showed depletion in metabolic pathways involved in biosynthesis of sulphur-containing amino acids. Only Group 2 showed significant shifts towards the donors' microbial composition [ADONIS: R2 = 0.15, p = 0.008] and significantly increased Eubacterium_sp_AF228LB post-intervention [β-coefficient 2.66, 95% confidence interval 2.1-3.3, q < 0.05] which was inversely correlated with faecal calprotectin [rho = -0.52, p = 0.035]. Moreover, pathways involved in gut inflammation and barrier function including branched chain amino acids were enriched post-intervention in Group 2 and were significantly inversely correlated with faecal calprotectin. CONCLUSION FMT from diet conditioned donors followed by the UCED led to microbial alterations associated with favourable microbial profiles which correlated with decreased faecal calprotectin. Our findings support further exploration of the additive benefit of dietary intervention for both donors and patients undergoing FMT as a potential treatment of UC.
Collapse
Affiliation(s)
- Haim Leibovitzh
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chen Sarbagili Shabat
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ayal Hirsch
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Zittan
- Gastroenterology Institute, IBD Unit, Haemek Medical Center, Afula, Israel
| | - Maria Chiara Mentella
- UOC di Nutrizione Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Petito
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Nathaniel Aviv Cohen
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Ron
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Naomi Fliss Isakov
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Department of Health, School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jorge Pfeffer
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Michal Yaakov
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon, Israel
| | - Caterina Fanali
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Laura Turchini
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Luca Masucci
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore - Fondazione Policlinico 'A. Gemelli' IRCSS, Rome, Italy
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore - Fondazione Policlinico 'A. Gemelli' IRCSS, Rome, Italy
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nitzan Kolonimos
- Gastroenterology Institute, IBD Unit, Haemek Medical Center, Afula, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Franco Scaldaferri
- Cemad [CENTER for Digestive Disease], UOC Medicina Internae Gastroenterologia, Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore- Fondazione Policlinico 'A. Gemelli' IRCCS, Rome, Italy
| | - Nitsan Maharshak
- Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Lewis G, Reczek S, Omozusi O, Hogue T, Cook MD, Hampton-Marcell J. Machine Learning Reveals Microbial Taxa Associated with a Swim across the Pacific Ocean. Biomedicines 2024; 12:2309. [PMID: 39457621 PMCID: PMC11504845 DOI: 10.3390/biomedicines12102309] [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: 08/29/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Purpose: This study aimed to characterize the association between microbial dynamics and excessive exercise. Methods: Swabbed fecal samples, body composition (percent body fat), and swimming logs were collected (n = 94) from a single individual over 107 days as he swam across the Pacific Ocean. The V4 region of the 16S rRNA gene was sequenced, generating 6.2 million amplicon sequence variants. Multivariate analysis was used to analyze the microbial community structure, and machine learning (random forest) was used to model the microbial dynamics over time using R statistical programming. Results: Our findings show a significant reduction in percent fat mass (Pearson; p < 0.01, R = -0.89) and daily swim distance (Spearman; p < 0.01, R = -0.30). Furthermore, the microbial community structure became increasingly similar over time (PERMANOVA; p < 0.01, R = -0.27). Decision-based modeling (random forest) revealed the genera Alistipes, Anaerostipes, Bifidobacterium, Butyricimonas, Lachnospira, Lachnobacterium, and Ruminococcus as important microbial biomarkers of excessive exercise for explaining variations observed throughout the swim (OOB; R = 0.893). Conclusions: We show that microbial community structure and composition accurately classify outcomes of excessive exercise in relation to body composition, blood pressure, and daily swim distance. More importantly, microbial dynamics reveal the microbial taxa significantly associated with increased exercise volume, highlighting specific microbes responsive to excessive swimming.
Collapse
Affiliation(s)
- Garry Lewis
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA; (G.L.); (S.R.)
| | - Sebastian Reczek
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA; (G.L.); (S.R.)
| | - Osayenmwen Omozusi
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Taylor Hogue
- Department of Kinesiology, North Carolina Agriculture and Technical State University, Greensboro, NC 27411, USA; (T.H.); (M.D.C.)
| | - Marc D. Cook
- Department of Kinesiology, North Carolina Agriculture and Technical State University, Greensboro, NC 27411, USA; (T.H.); (M.D.C.)
- Center of Integrative Health Disparities and Equity Research, North Carolina Agriculture and Technical State University, Greensboro, NC 27411, USA
| | - Jarrad Hampton-Marcell
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA; (G.L.); (S.R.)
| |
Collapse
|
29
|
Liu T, Ji H, Li Z, Luan Y, Zhu C, Li D, Gao Y, Yan Z. Gut microbiota causally impacts adrenal function: a two-sample mendelian randomization study. Sci Rep 2024; 14:23338. [PMID: 39375408 PMCID: PMC11458771 DOI: 10.1038/s41598-024-73420-w] [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: 03/27/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Some studies have reported that the gut microbiota can influence adrenal-related hormone levels. However, the causal effects of the gut microbiota on adrenal function remain unknown. Therefore, we employed a two-sample Mendelian randomization (MR) study to systematically investigate the impact of gut microbiota on the function of different regions of the adrenal gland. The summary statistics for gut microbiota and adrenal-related hormones used in the two-sample MR analysis were derived from publicly available genome-wide association studies (GWAS). In the MR analysis, inverse variance weighting (IVW) was used as the primary method, with MR-Egger, weighted median, and cML-MA serving as supplementary methods for causal inference. Sensitivity analyses such as the MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were used to assess pleiotropy and heterogeneity. We identified 27 causal relationships between 23 gut microbiota and adrenal function using the IVW method. Among these, Sellimonas enhanced the function of the adrenal cortex reticularis zone (beta = 0.008, 95% CI: 0.002-0.013, P = 0.0057). The cML-MA method supported our estimate (beta = 0.009, 95% CI: 0.004-0.013, P = 2 × 10- 4). Parasutterella, Sutterella, and Anaerofilum affect the functioning of different regions of the adrenal gland. Notably, pleiotropy was not observed. Our findings revealed that the gut microbiota is causally associated with adrenal function. This enhances our understanding of the gut-microbiota-brain axis and provides assistance in the early diagnosis and treatment of adrenal-related diseases in clinical practice.
Collapse
Affiliation(s)
- Tonghu Liu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Hongfei Ji
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhiyuan Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yongkun Luan
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Congcong Zhu
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Dongxiao Li
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Henan Children's Neurodevelopment Engineering Research Center, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China.
| | - Yukui Gao
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, 241001, Anhui, People's Republic of China.
| | - Zechen Yan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
- Henan Engineering Research Center of Tumor Molecular diagnosis and treatment, Zhengzhou, 450001, Henan, People's Republic of China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
30
|
Gilley SP, Ruebel ML, Chintapalli SV, Wright CJ, Rozance PJ, Shankar K. Calorie restriction during gestation impacts maternal and offspring fecal microbiome in mice. Front Endocrinol (Lausanne) 2024; 15:1423464. [PMID: 39429739 PMCID: PMC11487197 DOI: 10.3389/fendo.2024.1423464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 10/22/2024] Open
Abstract
Background Maternal undernutrition is the most common cause of fetal growth restriction (FGR) worldwide. FGR increases morbidity and mortality during infancy, as well as contributes to adult-onset diseases including obesity and type 2 diabetes. The role of the maternal or offspring microbiome in growth outcomes following FGR is not well understood. Methods FGR was induced by 30% maternal calorie restriction (CR) during the second half of gestation in C57BL/6 mice. Pup weights were obtained on day of life 0, 1, and 7 and ages 3, 4 and 16 weeks. Fecal pellets were collected from pregnant dams at gestational day 18.5 and from offspring at ages 3 and 4 weeks of age. Bacterial genomic DNA was used for amplification of the V4 variable region of the 16S rRNA gene. Multivariable associations between maternal CR and taxonomic abundance were assessed using the MaAsLin2 package. Associations between microbial taxa and offspring outcomes were performed using distance-based redundancy analysis and Pearson correlations. Results FGR pups weighed about 20% less than controls. Beta but not alpha diversity differed between control and CR dam microbiomes. CR dams had lower relative abundance of Turicibacter, Flexispira, and Rikenella, and increased relative abundance of Parabacteroides and Prevotella. Control and FGR offspring microbiota differed by beta diversity at ages 3 and 4 weeks. At 3 weeks, FGR offspring had decreased relative abundance of Akkermansia and Sutterella and increased relative abundance of Anaerostipes and Paraprevotella. At 4 weeks, FGR animals had decreased relative abundance of Allobaculum, Sutterella, Bifidobacterium, and Lactobacillus, among others, and increased relative abundance of Turcibacter, Dorea, and Roseburia. Maternal Helicobacter abundance was positively associated with offspring weight. Akkermansia abundance at age 3 and 4 weeks was negatively associated with adult weight. Conclusions We demonstrate gut microbial dysbiosis in pregnant dams and offspring at two timepoints following maternal calorie restriction. Additional research is needed to test for functional roles of the microbiome in offspring growth outcomes.
Collapse
Affiliation(s)
- Stephanie P. Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, United States
| | - Meghan L. Ruebel
- Microbiome and Metabolism Research Unit (MMRU), United States Department of Agriculture - Agricultural Research Service (USDA-ARS), Southeast Area, Little Rock, AR, United States
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sree V. Chintapalli
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Clyde J. Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Paul J. Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
31
|
Gurung M, Mulakala BK, Schlegel BT, Rajasundaram D, Shankar K, Bode L, Ruebel ML, Sims C, Martinez A, Andres A, Yeruva L. Maternal immune cell gene expression associates with maternal gut microbiome, milk composition and infant gut microbiome. Clin Nutr ESPEN 2024; 63:903-918. [PMID: 39209027 DOI: 10.1016/j.clnesp.2024.08.027] [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: 05/24/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Pre-pregnancy overweight and obesity promote deleterious health impacts on both mothers during pregnancy and the offspring. Significant changes in the maternal peripheral blood mononuclear cells (PBMCs) gene expression due to obesity are well-known. However, the impact of pre-pregnancy overweight on immune cell gene expression during pregnancy and its association with maternal and infant outcomes is not well explored. METHODS Blood samples were collected from healthy normal weight (NW, pre-pregnancy BMI 18.5-24.9) or overweight (OW, pre-pregnancy BMI 25-29.9) 2nd parity pregnant women at 12, 24 and 36 weeks of pregnancy. PBMCs were isolated from the blood and subjected to mRNA sequencing. Maternal and infant microbiota were analyzed by 16S rRNA gene sequencing. Integrative multi-omics data analysis was performed to evaluate the association of gene expression with maternal diet, gut microbiota, milk composition, and infant gut microbiota. RESULTS Gene expression analysis revealed that 453 genes were differentially expressed in the OW women compared to NW women at 12 weeks of pregnancy, out of which 354 were upregulated and 99 were downregulated. Several up-regulated genes in the OW group were enriched in inflammatory, chemokine-mediated signaling and regulation of interleukin-8 production-related pathways. At 36 weeks of pregnancy healthy eating index score was positively associated with several genes that include, DTD1, ELOC, GALNT8, ITGA6-AS1, KRT17P2, NPW, POT1-AS1 and RPL26. In addition, at 36 weeks of pregnancy, genes involved in adipocyte functions, such as NG2 and SMTNL1, were negatively correlated to human milk 2'FL and total fucosylated oligosaccharides content collected at 1 month postnatally. Furthermore, infant Akkermansia was positively associated with maternal PBMC anti-inflammatory genes that include CPS1 and RAB7B, at 12 and 36 weeks of pregnancy. CONCLUSIONS These findings suggest that prepregnancy overweight impacts the immune cell gene expression profile, particularly at 12 weeks of pregnancy. Furthermore, deciphering the complex association of PBMC's gene expression levels with maternal gut microbiome and milk composition and infant gut microbiome may aid in developing strategies to mitigate obesity-mediated effects.
Collapse
Affiliation(s)
- Manoj Gurung
- Microbiome and Metabolism Research Unit (MMRU), Southeast Area, USDA-ARS, Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Bharath Kumar Mulakala
- Microbiome and Metabolism Research Unit (MMRU), Southeast Area, USDA-ARS, Arkansas Children's Nutrition Center, Little Rock, AR, USA; Texas A & M, IHA, College Station, TX, USA
| | - Brent Thomas Schlegel
- University of Pittsburgh Medical Center (UPMC), Children's Hospital of Pittsburgh, PA, USA
| | - Dhivyaa Rajasundaram
- University of Pittsburgh Medical Center (UPMC), Children's Hospital of Pittsburgh, PA, USA
| | - Kartik Shankar
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lars Bode
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Meghan L Ruebel
- Microbiome and Metabolism Research Unit (MMRU), Southeast Area, USDA-ARS, Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Clark Sims
- Department of Pediatrics, University of Arkansas of Medical Sciences, Little Rock, AR, USA
| | - Audrey Martinez
- Department of Pediatrics, University of Arkansas of Medical Sciences, Little Rock, AR, USA
| | - Aline Andres
- Department of Pediatrics, University of Arkansas of Medical Sciences, Little Rock, AR, USA
| | - Laxmi Yeruva
- Microbiome and Metabolism Research Unit (MMRU), Southeast Area, USDA-ARS, Arkansas Children's Nutrition Center, Little Rock, AR, USA.
| |
Collapse
|
32
|
Zheng B, Xu J, Zhang Y, Qin J, Yuan D, Fan T, Wu W, Chen Y, Jiang Y. MBCN: A novel reference database for Effcient Metagenomic analysis of human gut microbiome. Heliyon 2024; 10:e37422. [PMID: 39315152 PMCID: PMC11417245 DOI: 10.1016/j.heliyon.2024.e37422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/09/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Metagenomic shotgun sequencing data can identify microbes and their proportions. But metagenomic shotgun data profiling results obtained from multiple projects using different reference databases are difficult to compare and apply meta-analysis. Our work aims to create a novel collection of human gut prokaryotic genomes, named Microbiome Collection Navigator (MBCN). 2379 human gut metagenomic samples are screened, and 16,785 metagenome-assembled genomes (MAGs) are assembled using a standardized pipeline. In addition, MAGs are combined with the representative genomes from public prokaryotic genomes collections to cluster, and pan-genomes for each cluster's genomes are constructed to build Kraken2 and Bracken databases. The databases built by MBCN are more comprehensive and accurate for profiling metagenomic reads comparing with other collections on simulated reads and virtual bio-projects. We profile 1082 human gut metagenomic samples with MBCN database and organize profiles and metadata on the web program. Meanwhile, using MBCN as a reference database, we also develop a unified, standardized, and systematic metagenomic analysis pipeline and platform, named MicrobiotaCN (http://www.microbiota.cn) and common statistical and visualization tools for microbiome research are integrated into the web program. Taken together, MBCN and MicrobiotaCN can be a valuable resource and a powerful tool that allows researchers to perform metagenomic analysis by a unified pipeline efficiently.
Collapse
Affiliation(s)
- Bo Zheng
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, PR China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, PR China
| | - Junming Xu
- Department of Human Microbiome, Promegene Institute, Shenzhen, 518000, PR China
| | - Yijie Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, PR China
| | - Junjie Qin
- Department of Human Microbiome, Promegene Institute, Shenzhen, 518000, PR China
| | - Decai Yuan
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, PR China
| | - Tingting Fan
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, PR China
| | - Weibin Wu
- Shenzhen Bay Biotechnology Co., Ltd. Shenzhen, 518110, PR China
| | - Yan Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, PR China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, 518055, PR China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, PR China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China
| |
Collapse
|
33
|
Chen J, Wang Y, Yao H, Li Y, Song H. Uncovering a Causal Connection between Gut Microbiota and Six Thyroid Diseases: A Two-Sample Mendelian Randomization Study. BIOLOGY 2024; 13:714. [PMID: 39336141 PMCID: PMC11428278 DOI: 10.3390/biology13090714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Recent studies have established associations between the gut microbiota (GM) and thyroid diseases (TDs). However, their causal relationships remain elusive. METHODS To investigate this causality, we conducted a two-sample Mendelian randomization (MR) analysis using genome-wide association study (GWAS) data from MiBioGen and FinnGen, with GM as the exposure and six TDs as outcomes. RESULTS We identified 32 microbial taxa linked to the risk of six TDs. The Clostridium innocuum group, Ruminiclostridium5, and Lachnoclostridium exhibited protective effects against nontoxic diffuse goiter (NDG). Conversely, an increased risk of NDG was associated with Ruminococcaceae UCG002, Alistipes, Methanobrevibacter, Marvinbryantia, and Ruminococcaceae UCG014. Bifidobacterium and Sutterella were protective against nontoxic multinodular goiter (NMG), while the Ruminococcus gauvreauii group and Rikenellaceae RC9 gut group heightened NMG risk. Protective effects against nontoxic single thyroid nodule (NSTN) were observed with Defluviitaleaceae UCG011, Ruminococcus1, and Ruminococcaceae UCG010, whereas increased risk was linked to Alistipes, the Ruminococcus gauvreauii group, and Lachnospiraceae UCG010. Ruminiclostridium9, Victivallis, and Butyricimonas offered protection against thyrotoxicosis with Graves' Disease (GD), while the Eubacterium rectale group, Desulfovibrio, Bifidobacterium, Collinsella, Oscillospira, and Catenibacterium were risk factors. For thyrotoxicosis with Plummer Disease (PD), protective taxa included Butyricimonas and Lachnospira, whereas Dorea, Eggerthella, Odoribacter, Lactobacillus, Intestinimonas, and Phascolarctobacterium increased risk. Lastly, Parasutterella was protective against thyrotoxicosis with toxic single thyroid nodule (TSTN), while increased risk was associated with Sutterella, Oscillibacter, and Clostridium sensu stricto1. CONCLUSIONS Our findings support a causal relationship between specific GM and TDs at the genetic level, laying the foundation for future research into potential mechanisms and the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Jiahao Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Yu Wang
- Graduate School of Jiangxi, University of Chinese Medicine, Nanchang 330004, China; (Y.W.); (Y.L.)
| | - Hang Yao
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai 264003, China;
| | - Yuxin Li
- Graduate School of Jiangxi, University of Chinese Medicine, Nanchang 330004, China; (Y.W.); (Y.L.)
| | - Hong Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| |
Collapse
|
34
|
Heil BA, van Heule M, Thompson SK, Kearns TA, Beckers KF, Oberhaus EL, King G, Daels P, Dini P, Sones JL. Metagenomic characterization of the equine endometrial microbiome during anestrus. J Equine Vet Sci 2024; 140:105134. [PMID: 38909766 DOI: 10.1016/j.jevs.2024.105134] [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: 12/31/2023] [Revised: 05/17/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
The equine uterus is highly interrogated during estrus prior to breeding and establishing pregnancy. Many studies in mares have been performed during estrus under the influence of high estrogen concentrations, including the equine estrual microbiome. To date, it is unknown how the uterine microbiome of the mare is influenced by cyclicity; while, the equine vaginal microbiome is stable throughout the estrous cycle. We hypothesized that differences would exist between the equine endometrial microbiome of mares in estrus and anestrus. The aim of this study was two-fold: to characterize the resident endometrial microbiome of healthy mares during anestrus and to compare this with estrus. Double-guarded endometrial swabs were taken from healthy mares during estrus (n = 16) and in the following non-breeding season during anestrus (n = 8). Microbial population was identified using 16S rRNA sequencing. Our results suggest that the equine uterine microbiome in estrus has a low diversity and low richness, while during anestrus, a higher diversity and higher richness were seen compared to estrus. Despite this difference, both the estrus and anestrus endometrial microbiome were dominated by Proteobacteria, Firmicutes, and Bacteroidota. The composition of the microbial community between anestrus and estrus was significantly different. This may be explained by the difference in the composition of the endometrial immune milieu based on the stage of the cycle. Further research investigating the function of the equine endometrial microbiome and dynamics changes within the uterine environment is required.
Collapse
Affiliation(s)
- B A Heil
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - M van Heule
- Department of Population Health and Reproduction (PHR), School of Veterinary Medicine, UCDavis, Davis, CA, 95616, USA; Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, UGent, Merelbeke, 9820, Belgium
| | - S K Thompson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - T A Kearns
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - K F Beckers
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - E L Oberhaus
- School of Animal Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - G King
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - P Daels
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, UGent, Merelbeke, 9820, Belgium
| | - P Dini
- Department of Population Health and Reproduction (PHR), School of Veterinary Medicine, UCDavis, Davis, CA, 95616, USA
| | - J L Sones
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA; Equine Reproduction Laboratory, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80521, USA.
| |
Collapse
|
35
|
Aldegheri L, Kharrat F, Conti A, Monica F, Busa F, Campisciano G, Zanotta N, Cason C, Comar M. Impact of Human Milk Oligosaccharides and Probiotics on Gut Microbiome and Mood in Autism: A Case Report. Microorganisms 2024; 12:1625. [PMID: 39203467 PMCID: PMC11356532 DOI: 10.3390/microorganisms12081625] [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: 07/22/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Recent evidence has highlighted the role of the gut-brain axis in the progression of autism spectrum disorder (ASD), with significant changes in the gut microbiome of individuals with this condition. This report investigates the effects of probiotics and human milk oligosaccharide (HMO) supplements on the gut microbiome, inflammatory cytokine profile, and clinical outcomes in an ASD adolescent with chronic gastrointestinal dysfunction and cognitive impairment. Following treatment, we observed a decrease in proinflammatory cytokines' concentration alongside Sutterella relative abundance, a bacterium reported to be linked with gastrointestinal diseases. Also, we reported a notable increase in mood stability. The study aims to evaluate the use of gut microbiome-based therapy in selected ASD patients, highlighting its potential to improve related clinical symptoms.
Collapse
Affiliation(s)
- Luana Aldegheri
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Feras Kharrat
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Andrea Conti
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Fabio Monica
- Department of Gastroenterology and Endoscopy, Trieste University Hospital, Strada di Fiume 447, 34149 Trieste, Italy;
| | | | - Giuseppina Campisciano
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Nunzia Zanotta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Carolina Cason
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (L.A.); (F.K.); (A.C.); (G.C.); (N.Z.); (C.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| |
Collapse
|
36
|
Li Q, Sun Y, Zhai K, Geng B, Dong Z, Ji L, Chen H, Cui Y. Microbiota-induced inflammatory responses in bladder tumors promote epithelial-mesenchymal transition and enhanced immune infiltration. Physiol Genomics 2024; 56:544-554. [PMID: 38808774 DOI: 10.1152/physiolgenomics.00032.2024] [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: 03/25/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 05/30/2024] Open
Abstract
The intratumoral microbiota can modulate the tumor immune microenvironment (TIME); however, the underlying mechanism by which intratumoral microbiota influences the TIME in urothelial carcinoma of the bladder (UCB) remains unclear. To address this, we collected samples from 402 patients with UCB, including paired host transcriptome and tumor microbiome data, from The Cancer Genome Atlas (TCGA). We found that the intratumoral microbiome profiles were significantly correlated with the expression pattern of epithelial-mesenchymal transition (EMT)-related genes. Furthermore, we detected that the genera Lachnoclostridium and Sutterella in tumors could indirectly promote the EMT program by inducing an inflammatory response. Moreover, the inflammatory response induced by these two intratumoral bacteria further enhanced intratumoral immune infiltration, affecting patient survival and response to immunotherapy. In addition, an independent immunotherapy cohort of 348 patients with bladder cancer was used to validate our results. Collectively, our study elucidates the potential mechanism by which the intratumoral microbiota influences the TIME of UCB and provides a new guiding strategy for the targeted therapy of UCB.NEW & NOTEWORTHY The intratumoral microbiota may mediate the bladder tumor inflammatory response, thereby promoting the epithelial-mesenchymal transition program and influencing tumor immune infiltration.
Collapse
Affiliation(s)
- Qiang Li
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Yichao Sun
- Department of Operating Room, Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Kun Zhai
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Bingzhi Geng
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Zhenkun Dong
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing, People's Republic of China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, People's Republic of China
| | - Hui Chen
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Yan Cui
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| |
Collapse
|
37
|
Huang C, Liu D, Yang S, Huang Y, Wei X, Zhang P, Lin J, Xu B, Liu Y, Guo D, Li Y, Li J, Zhang H. Effect of time-restricted eating regimen on weight loss is mediated by gut microbiome. iScience 2024; 27:110202. [PMID: 38993674 PMCID: PMC11238135 DOI: 10.1016/j.isci.2024.110202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/16/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
Time-restricted eating (TRE) is a promising obesity management strategy, but weight-loss efficacy varies among participants, and the underlying mechanism is unclear. The study aimed to investigate the role of gut microbiota in weight-loss response during long-term TRE intervention. We analyzed data from 51 obese adults in a 12-month TRE program, categorizing them into distinct weight loss groups (DG) and moderate weight loss groups (MG) based on their TRE responses. Shotgun metagenomic sequencing analysis revealed a significant increase in species closely associated with weight loss effectiveness and metabolic parameter changes in the DG group. Pathways related to fatty acid biosynthesis, glycogen biosynthesis, and nucleotide metabolism were reduced in the DG group and enhanced in the MG group. Next, we identified nine specific species at baseline that contributed better responses to TRE intervention and significant weight loss. Collectively, gut microbiota contributes to responsiveness heterogeneity in TRE and can predict weight-loss effectiveness.
Collapse
Affiliation(s)
- Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shunyu Yang
- Department of Nutrition, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yating Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan 030000, Shanxi, China
- Core Laboratory, Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University; Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan 030000, Shanxi, China
| | - Jin Li
- Department of Endocrinology and Metabolism, Shan Xi Medical University Second Hospital, Shan Xi Medical University, Taiyuan 030000, Shanxi, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| |
Collapse
|
38
|
Rodriguez CI, Isobe K, Martiny JBH. Short-term dietary fiber interventions produce consistent gut microbiome responses across studies. mSystems 2024; 9:e0013324. [PMID: 38742890 PMCID: PMC11237734 DOI: 10.1128/msystems.00133-24] [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: 01/29/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
The composition of the human gut microbiome varies tremendously among individuals, making the effects of dietary or treatment interventions difficult to detect and characterize. The consumption of fiber is important for gut health, yet the specific effects of increased fiber intake on the gut microbiome vary across studies. The variation in study outcomes might be due to inter-individual (or inter-population) variation or to the details of the interventions including the types of fiber, length of study, size of cohort, and molecular approaches. Thus, to identify generally (on average) consistent fiber-induced responses in the gut microbiome of healthy individuals, we re-analyzed 16S rRNA sequencing data from 21 dietary fiber interventions from 12 human studies, which included 2,564 fecal samples from 538 subjects across all interventions. Short-term increases in dietary fiber consumption resulted in highly consistent gut bacterial community responses across studies. Increased fiber consumption explained an average of 1.5% of compositional variation (vs 82% of variation attributed to the individual), reduced alpha-diversity, and resulted in phylogenetically conserved responses in relative abundances among bacterial taxa. Additionally, we identified bacterial clades, at approximately the genus level, that were highly consistent in their response (on average, increasing or decreasing in their relative abundance) to dietary fiber interventions across the studies. IMPORTANCE Our study is an example of the power of synthesizing and reanalyzing 16S rRNA microbiome data from many intervention studies. Despite high inter-individual variation of the composition of the human gut microbiome, dietary fiber interventions cause a consistent response both in the degree of change and the particular taxa that respond to increased fiber.
Collapse
Affiliation(s)
- Cynthia I. Rodriguez
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, USA
| | - Kazuo Isobe
- Institute of Ecology, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Jennifer B. H. Martiny
- Department of Ecology and Evolutionary Biology, University of California, Irvine, California, USA
| |
Collapse
|
39
|
Lee S, Tejesvi MV, Hurskainen E, Aasmets O, Plaza-Díaz J, Franks S, Morin-Papunen L, Tapanainen JS, Ruuska TS, Altmäe S, Org E, Salumets A, Arffman RK, Piltonen TT. Gut bacteriome and mood disorders in women with PCOS. Hum Reprod 2024; 39:1291-1302. [PMID: 38614956 PMCID: PMC11145006 DOI: 10.1093/humrep/deae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/19/2024] [Indexed: 04/15/2024] Open
Abstract
STUDY QUESTION How does the gut bacteriome differ based on mood disorders (MDs) in women with polycystic ovary syndrome (PCOS), and how can the gut bacteriome contribute to the associations between these two conditions? SUMMARY ANSWER Women with PCOS who also have MDs exhibited a distinct gut bacteriome with reduced alpha diversity and a significantly lower abundance of Butyricicoccus compared to women with PCOS but without MDs. WHAT IS KNOWN ALREADY Women with PCOS have a 4- to 5-fold higher risk of having MDs compared to women without PCOS. The gut bacteriome has been suggested to influence the pathophysiology of both PCOS and MDs. STUDY DESIGN, SIZE, DURATION This population-based cohort study was derived from the Northern Finland Birth Cohort 1966 (NFBC1966), which includes all women born in Northern Finland in 1966. Women with PCOS who donated a stool sample at age 46 years (n = 102) and two BMI-matched controls for each case (n = 205), who also responded properly to the MD criteria scales, were included. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 102 women with PCOS and 205 age- and BMI-matched women without PCOS were included. Based on the validated MD criteria, the subjects were categorized into MD or no-MD groups, resulting in the following subgroups: PCOS no-MD (n = 84), PCOS MD (n = 18), control no-MD (n = 180), and control MD (n = 25). Clinical characteristics were assessed at age 31 years and age 46 years, and stool samples were collected from the women at age 46 years, followed by the gut bacteriome analysis using 16 s rRNA sequencing. Alpha diversity was assessed using observed features and Shannon's index, with a focus on genera, and beta diversity was characterized using principal components analysis (PCA) with Bray-Curtis Dissimilarity at the genus level. Associations between the gut bacteriome and PCOS-related clinical features were explored by Spearman's correlation coefficient. A P-value for multiple testing was adjusted with the Benjamini-Hochberg false discovery rate (FDR) method. MAIN RESULTS AND THE ROLE OF CHANCE We observed changes in the gut bacteriome associated with MDs, irrespective of whether the women also had PCOS. Similarly, PCOS MD cases showed a lower alpha diversity (Observed feature, PCOS no-MD, median 272; PCOS MD, median 208, FDR = 0.01; Shannon, PCOS no-MD, median 5.95; PCOS MD, median 5.57, FDR = 0.01) but also a lower abundance of Butyricicoccus (log-fold changeAnalysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC)=-0.90, FDRANCOM-BC=0.04) compared to PCOS no-MD cases. In contrast, in the controls, the gut bacteriome did not differ based on MDs. Furthermore, in the PCOS group, Sutterella showed positive correlations with PCOS-related clinical parameters linked to obesity (BMI, r2=0.31, FDR = 0.01; waist circumference, r2=0.29, FDR = 0.02), glucose metabolism (fasting glucose, r2=0.46, FDR < 0.001; fasting insulin, r2=0.24, FDR = 0.05), and gut barrier integrity (zonulin, r2=0.25, FDR = 0.03). LIMITATIONS, REASONS FOR CAUTION Although this was the first study to assess the link between the gut bacteriome and MDs in PCOS and included the largest PCOS dataset for the gut microbiome analysis, the number of subjects stratified by the presence of MDs was limited when contrasted with previous studies that focused on MDs in a non-selected population. WIDER IMPLICATIONS OF THE FINDINGS The main finding is that gut bacteriome is associated with MDs irrespective of the PCOS status, but PCOS may also modulate further the connection between the gut bacteriome and MDs. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the European Union's Horizon 2020 Research and Innovation Programme under the Marie Sklodowska-Curie Grant Agreement (MATER, No. 813707), the Academy of Finland (project grants 315921, 321763, 336449), the Sigrid Jusélius Foundation, Novo Nordisk Foundation (NNF21OC0070372), grant numbers PID2021-12728OB-100 (Endo-Map) and CNS2022-135999 (ROSY) funded by MCIN/AEI/10.13039/501100011033 and ERFD A Way of Making Europe. The study was also supported by EU QLG1-CT-2000-01643 (EUROBLCS) (E51560), NorFA (731, 20056, 30167), USA/NIH 2000 G DF682 (50945), the Estonian Research Council (PRG1076, PRG1414), EMBO Installation (3573), and Horizon 2020 Innovation Grant (ERIN, No. EU952516). The funders did not participate in any process of the study. We have no conflicts of interest to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- S Lee
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - M V Tejesvi
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Ecology and Genetics, University of Oulu, Oulu, Finland
| | - E Hurskainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - O Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - J Plaza-Díaz
- Faculty of Pharmacy, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - S Franks
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - L Morin-Papunen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - J S Tapanainen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR—Cantonal Hospital of and University of Fribourg, Fribourg, Switzerland
| | - T S Ruuska
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - S Altmäe
- Faculty of Pharmacy, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - E Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - A Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| | - R K Arffman
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - T T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
40
|
Pederzoli F, Riba M, Venegoni C, Marandino L, Bandini M, Alchera E, Locatelli I, Raggi D, Giannatempo P, Provero P, Lazarevic D, Moschini M, Lucianò R, Gallina A, Briganti A, Montorsi F, Salonia A, Necchi A, Alfano M. Stool Microbiome Signature Associated with Response to Neoadjuvant Pembrolizumab in Patients with Muscle-invasive Bladder Cancer. Eur Urol 2024; 85:417-421. [PMID: 38184414 DOI: 10.1016/j.eururo.2023.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 11/18/2023] [Accepted: 12/22/2023] [Indexed: 01/08/2024]
Abstract
Neoadjuvant pembrolizumab has been shown to be a valid treatment for patients affected by muscle-invasive bladder cancer (MIBC), as demonstrated in the PURE-01 clinical trial (NCT02736266). Among the tumor-extrinsic factors influencing immunotherapy efficacy, extensive data highlighted that the microbiome is a central player in immune-mediated anticancer activity. This report aimed to investigate the composition and role of stool microbiome in patients enrolled in the PURE-01 clinical trial. An orthotopic animal model of bladder cancer (MB49-Luc) was used to support some of the findings from human data. An analysis of stool microbiome before pembrolizumab was conducted for 42 patients, of whom 23 showed a pathologic response. The information in the preclinical model of orthotopic bladder cancer treated with anti-PD-1 antibody or control isotype was validated. Linear discriminant analysis effect size and linear models were used to identify the bacterial taxa enriched in either responders or nonresponders. The identified taxa were also tested for their association with event-free survival (EFS). Survival at 31 d after tumor instillation was used as the study endpoint in the preclinical model. Responders and nonresponders emerged to differ in terms of enrichment for 16 bacterial taxa. Of these, the genus Sutterella was enriched in responders, while the species Ruminococcus bromii was enriched in nonresponders. The negative impact of R. bromii on anti-PD-1 antibody activity was also observed in the preclinical model. EFS and survival of the preclinical model showed a negative role of R. bromii. We found different stool bacterial taxa associated with the response or lack of response to neoadjuvant pembrolizumab. Moreover, we provided experimental data about the negative role of R. bromii on immunotherapy response. Further studies are needed to externally validate our findings and provide mechanistic insights about the host-pathogen interactions in MIBC. PATIENT SUMMARY: Using prepembrolizumab stool samples collected from patients enrolled in the PURE-01 clinical trials, we identified some bacterial taxa that were enriched in patients who either responded or did not respond to immunotherapy. Using an animal model of bladder cancer, we gathered further evidence of the negative impact of the Ruminococcus bromii on immunotherapy efficacy. Further studies are needed to confirm the current findings and test the utility of these bacteria as predictive markers of immunotherapy response.
Collapse
Affiliation(s)
- Filippo Pederzoli
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Michela Riba
- Center for Omics Sciences, IRCCS San Raffaele Hospital, Milano, Italy
| | - Chiara Venegoni
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Laura Marandino
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Bandini
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Elisa Alchera
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Irene Locatelli
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Daniele Raggi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Paolo Provero
- Center for Omics Sciences, IRCCS San Raffaele Hospital, Milano, Italy; Department of Neurosciences 'Rita Levi Montalcini', University of Turin, Turin, Italy
| | - Dejan Lazarevic
- Center for Omics Sciences, IRCCS San Raffaele Hospital, Milano, Italy
| | - Marco Moschini
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Roberta Lucianò
- Department of Anatomic Pathology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Andrea Gallina
- Istituto Oncologico della Svizzera Italiana (IOSI), Bellinzona, Switzerland
| | - Alberto Briganti
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Francesco Montorsi
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Salonia
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Necchi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Massimo Alfano
- Unit of Urology, Division of Experimental Oncology, URI, IRCCS San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
41
|
Baltazar-Díaz TA, Andrade-Villanueva JF, Sánchez-Álvarez P, Amador-Lara F, Holguín-Aguirre T, Sánchez-Reyes K, Álvarez-Zavala M, López-Roa RI, Bueno-Topete MR, González-Hernández LA. A Two-Faced Gut Microbiome: Butyrogenic and Proinflammatory Bacteria Predominate in the Intestinal Milieu of People Living with HIV from Western Mexico. Int J Mol Sci 2024; 25:4830. [PMID: 38732048 PMCID: PMC11084381 DOI: 10.3390/ijms25094830] [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: 02/29/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/13/2024] Open
Abstract
HIV infection results in marked alterations in the gut microbiota (GM), such as the loss of microbial diversity and different taxonomic and metabolic profiles. Despite antiretroviral therapy (ART) partially ablating gastrointestinal alterations, the taxonomic profile after successful new ART has shown wide variations. Our objective was to determine the GM composition and functions in people living with HIV (PLWHIV) under ART in comparison to seronegative controls (SC). Fecal samples from 21 subjects (treated with integrase strand-transfer inhibitors, INSTIs) and 18 SC were included. We employed 16S rRNA amplicon sequencing, coupled with PICRUSt2 and fecal short-chain fatty acid (SCFA) quantification by gas chromatography. The INSTI group showed a decreased α-diversity (p < 0.001) compared to the SC group, at the expense of increased amounts of Pseudomonadota (Proteobacteria), Segatella copri, Lactobacillus, and Gram-negative bacteria. Concurrently, we observed an enrichment in Megasphaera and Butyricicoccus, both SCFA-producing bacteria, and significant elevations in fecal butyrate in this group (p < 0.001). Interestingly, gut dysbiosis in PLWHIV was characterized by a proinflammatory environment orchestrated by Pseudomonadota and elevated levels of butyrate associated with bacterial metabolic pathways, as well as the evident presence of butyrogenic bacteria. The role of this unique GM in PLWHIV should be evaluated, as well as the use of butyrate-based supplements and ART regimens that contain succinate, such as tenofovir disoproxil succinate. This mixed profile is described for the first time in PLWHIV from Mexico.
Collapse
Affiliation(s)
- Tonatiuh Abimael Baltazar-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico;
- Instituto de Investigación en Inmunodeficiencias y VIH, Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Hospital 278, Guadalajara 44280, Mexico; (J.F.A.-V.); (K.S.-R.); (M.Á.-Z.)
| | - Jaime F. Andrade-Villanueva
- Instituto de Investigación en Inmunodeficiencias y VIH, Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Hospital 278, Guadalajara 44280, Mexico; (J.F.A.-V.); (K.S.-R.); (M.Á.-Z.)
- Unidad de VIH, Hospital Civil de Guadalajara Fray Antonio Alcalde, Hospital 278, Guadalajara 44280, Mexico; (P.S.-Á.); (F.A.-L.); (T.H.-A.)
| | - Paulina Sánchez-Álvarez
- Unidad de VIH, Hospital Civil de Guadalajara Fray Antonio Alcalde, Hospital 278, Guadalajara 44280, Mexico; (P.S.-Á.); (F.A.-L.); (T.H.-A.)
| | - Fernando Amador-Lara
- Unidad de VIH, Hospital Civil de Guadalajara Fray Antonio Alcalde, Hospital 278, Guadalajara 44280, Mexico; (P.S.-Á.); (F.A.-L.); (T.H.-A.)
| | - Tania Holguín-Aguirre
- Unidad de VIH, Hospital Civil de Guadalajara Fray Antonio Alcalde, Hospital 278, Guadalajara 44280, Mexico; (P.S.-Á.); (F.A.-L.); (T.H.-A.)
| | - Karina Sánchez-Reyes
- Instituto de Investigación en Inmunodeficiencias y VIH, Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Hospital 278, Guadalajara 44280, Mexico; (J.F.A.-V.); (K.S.-R.); (M.Á.-Z.)
| | - Monserrat Álvarez-Zavala
- Instituto de Investigación en Inmunodeficiencias y VIH, Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Hospital 278, Guadalajara 44280, Mexico; (J.F.A.-V.); (K.S.-R.); (M.Á.-Z.)
| | - Rocío Ivette López-Roa
- Laboratorio de Investigación y Desarrollo Farmacéutico, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Marcelino García Barragán 1421, Guadalajara 44430, Mexico;
| | - Miriam Ruth Bueno-Topete
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Guadalajara 44340, Mexico;
| | - Luz Alicia González-Hernández
- Instituto de Investigación en Inmunodeficiencias y VIH, Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Hospital 278, Guadalajara 44280, Mexico; (J.F.A.-V.); (K.S.-R.); (M.Á.-Z.)
- Unidad de VIH, Hospital Civil de Guadalajara Fray Antonio Alcalde, Hospital 278, Guadalajara 44280, Mexico; (P.S.-Á.); (F.A.-L.); (T.H.-A.)
| |
Collapse
|
42
|
Deng Z, Liu L, Liu W, Liu R, Ma T, Xin Y, Xie Y, Zhang Y, Zhou Y, Tang Y. Alterations in the fecal microbiota of methamphetamine users with bad sleep quality during abstinence. BMC Psychiatry 2024; 24:324. [PMID: 38664669 PMCID: PMC11046801 DOI: 10.1186/s12888-024-05773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Methamphetamine (MA) abuse has resulted in a plethora of social issues. Sleep disturbance is a prominent issue about MA addiction, which serve as a risk factor for relapse, and the gut microbiota could play an important role in the pathophysiological mechanisms of sleep disturbances. Therefore, improving sleep quality can be beneficial for treating methamphetamine addiction, and interventions addressing the gut microbiota may represent a promising approach. METHOD We recruited 70 MA users to investigate the associations between sleep quality and fecal microbiota by the Pittsburgh Sleep Quality Index (PSQI), which was divided into MA-GS (PSQI score < 7, MA users with good sleep quality, n = 49) and MA-BS group (PSQI score ≥ 7, MA users with bad sleep quality, n = 21). In addition, we compared the gut microbiota between the MA-GS and healthy control (HC, n = 38) groups. 16S rRNA sequencing was applied to identify the gut bacteria. RESULT The study revealed that the relative abundances of the Thermoanaerobacterales at the order level differed between the MA-GS and MA-BS groups. Additionally, a positive correlation was found between the relative abundance of the genus Sutterella and daytime dysfunction. Furthermore, comparisons between MA users and HCs revealed differences in beta diversity and relative abundances of various bacterial taxa. CONCLUSION In conclusion, the study investigated alterations in the gut microbiota among MA users. Furthermore, we demonstrated that the genus Sutterella changes may be associated with daytime dysfunction, suggesting that the genus Sutterella may be a biomarker for bad sleep quality in MA users.
Collapse
Affiliation(s)
- Zijing Deng
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Linzi Liu
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Wen Liu
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Ruina Liu
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shanxi, PR China
| | - Tao Ma
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Yide Xin
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Yu Xie
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Yifan Zhang
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Yifang Zhou
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China
| | - Yanqing Tang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, PR China.
| |
Collapse
|
43
|
Díez-Sánchez A, Lindholm HT, Vornewald PM, Ostrop J, Yao R, Single AB, Marstad A, Parmar N, Shaw TN, Martín-Alonso M, Oudhoff MJ. LSD1 drives intestinal epithelial maturation and controls small intestinal immune cell composition independent of microbiota in a murine model. Nat Commun 2024; 15:3412. [PMID: 38649356 PMCID: PMC11035651 DOI: 10.1038/s41467-024-47815-2] [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: 09/10/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Postnatal development of the gastrointestinal tract involves the establishment of the commensal microbiota, the acquisition of immune tolerance via a balanced immune cell composition, and maturation of the intestinal epithelium. While studies have uncovered an interplay between the first two, less is known about the role of the maturing epithelium. Here we show that intestinal-epithelial intrinsic expression of lysine-specific demethylase 1A (LSD1) is necessary for the postnatal maturation of intestinal epithelium and maintenance of this developed state during adulthood. Using microbiota-depleted mice, we find plasma cells, innate lymphoid cells (ILCs), and a specific myeloid population to depend on LSD1-controlled epithelial maturation. We propose that LSD1 controls the expression of epithelial-derived chemokines, such as Cxcl16, and that this is a mode of action for this epithelial-immune cell interplay in local ILC2s but not ILC3s. Together, our findings suggest that the maturing epithelium plays a dominant role in regulating the local immune cell composition, thereby contributing to gut homeostasis.
Collapse
Affiliation(s)
- Alberto Díez-Sánchez
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Håvard T Lindholm
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Pia M Vornewald
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jenny Ostrop
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rouan Yao
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andrew B Single
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anne Marstad
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Naveen Parmar
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tovah N Shaw
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mara Martín-Alonso
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Menno J Oudhoff
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
- Department of Health Sciences, Carleton University, Ottawa, Ontario, ON, Canada.
| |
Collapse
|
44
|
Qin P, He Y, Shao H, Jiang D. Genetic insights into gut microbiota and risk of prostatitis: a Mendelian randomization study. Front Microbiol 2024; 15:1389715. [PMID: 38680919 PMCID: PMC11045958 DOI: 10.3389/fmicb.2024.1389715] [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: 02/22/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Background The dysbiosis of gut microbiota (GM) is considered a contributing factor to prostatitis, yet the causality remains incompletely understood. Methods The genome-wide association study (GWAS) data for GM and prostatitis were sourced from MiBioGen and FinnGen R10, respectively. In the two-sample Mendelian randomization (MR) analysis, inverse variance weighting (IVW), MR-Egger, weighted median, simple mode, weighted mode, and maximum likelihood (ML) methods were utilized to investigate the causal relationship between GM and prostatitis. A series of sensitivity analysis were conducted to confirm the robustness of the main results obtained from the MR analysis. Results According to the IVW results, genus Sutterella (OR: 1.37, 95% CI: 1.09-1.71, p = 0.006) and genus Holdemania (OR: 1.21, 95% CI: 1.02-1.43, p = 0.028) were associated with an increased risk of prostatitis. The phylum Verrucomicrobia (OR: 0.76, 95% CI: 0.58-0.98, p = 0.033) and genus Parasutterella (OR: 0.84, 95% CI: 0.70-1.00, p = 0.045) exhibited a negative association with prostatitis, indicating a potential protective effect. Sensitivity analysis showed that these results were not affected by heterogeneity and horizontal pleiotropy. Furthermore, the majority of statistical methods yielded results consistent with those of the IVW analysis. Conclusions In this study, we identified two GM taxon that might be protective against prostatitis and two GM taxon that could increase the risk of developing prostatitis. These findings could potentially provide a valuable theoretical basis for the future development of preventive and therapeutic strategies for prostatitis.
Collapse
Affiliation(s)
| | | | | | - Dawei Jiang
- Department of Urology, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Jiaxing, China
| |
Collapse
|
45
|
Yu SY, Oh BS, Ryu SW, Bak JE, Heo ES, Moon JC, Jeong JH, Lee JH. Novel microbiota Mesosutterella faecium sp. nov. has a protective effect against inflammatory bowel disease. Front Microbiol 2024; 15:1342098. [PMID: 38633706 PMCID: PMC11022602 DOI: 10.3389/fmicb.2024.1342098] [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: 11/21/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
A novel Gram-negative, obligate anaerobe, non-motile, flagella-lacking, catalase- and oxidase-negative, coccobacilli-shaped bacterial strain designated AGMB02718T was isolated from swine feces. The 16S rRNA gene analysis indicated that strain AGMB02718T belonged to the genus Mesosutterella with the highest similarity to M. multiformis 4NBBH2T (= DSM 106860T) (sequence similarity of 96.2%), forming a distinct phylogenetic lineage. Its growth occurred at 25-45°C (optimal 37°C) and in 0.5-1% NaCl (optimal 0.5%). Strain AGMB02718T was asaccharolytic and contained menaquinone 6 (MK-6) and methylmenaquinone 6 (MMK-6) as the predominant respiratory quinones. The major cellular fatty acids in the isolate were C18:1ω9c and C16:0. Based on the whole-genome sequencing analysis, strain AGMB02718T had a 2,606,253 bp circular chromosome with a G + C content of 62.2%. The average nucleotide identity value between strain AGMB02718T and M. multiformis 4NBBH2T was 72.1%, while the digital DNA-DNA hybridization value was 20.9%. Interestingly, genome analysis suggested that strain AGMB02718T possessed a low-toxicity lipopolysaccharide (LPS) because the genome of the isolate does not include lpxJ and lpxM genes for Kdo2-Lipid A (KLA) assembly, which confers high toxicity to LPS. Moreover, in vitro macrophage stimulation assay confirmed that AGMB02718T produced LPS with low toxicity. Because the low-toxicity LPS produced by the Sutterellaceae family is involved in regulating host immunity and low-toxicity LPS-producing strains can help maintain host immune homeostasis, we evaluated the anti-inflammatory activity of strain AGMB02718T against inflammatory bowel disease (IBD). As a result, strain AGMB02718T was able to prevent the inflammatory response in a dextran sulfate sodium (DSS)-induced colitis model. Therefore, this strain represents a novel species of Mesosutterella that has a protective effect against DSS-induced colitis, and the proposed name is Mesosutterella faecium sp. nov. The type strain is AGMB02718T (=GDMCC 1.2717T = KCTC 25541T).
Collapse
Affiliation(s)
- Seung Yeob Yu
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Republic of Korea
| | - Byeong Seob Oh
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Seoung Woo Ryu
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Republic of Korea
| | - Jeong Eun Bak
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Republic of Korea
| | - Eun Seo Heo
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| | | | - Jae-Ho Jeong
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Hwasun, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
- University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
46
|
Liu Y, Zhu Q, Guo G, Xie Z, Li S, Lai C, Wu Y, Wang L, Zhong S. Causal associations of genetically predicted gut microbiota and blood metabolites with inflammatory states and risk of infections: a Mendelian randomization analysis. Front Microbiol 2024; 15:1342653. [PMID: 38585702 PMCID: PMC10995310 DOI: 10.3389/fmicb.2024.1342653] [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: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 04/09/2024] Open
Abstract
Background Inflammation serves as a key pathologic mediator in the progression of infections and various diseases, involving significant alterations in the gut microbiome and metabolism. This study aims to probe into the potential causal relationships between gut microbial taxa and human blood metabolites with various serum inflammatory markers (CRP, SAA1, IL-6, TNF-α, WBC, and GlycA) and the risks of seven common infections (gastrointestinal infections, dysentery, pneumonia, bacterial pneumonia, bronchopneumonia and lung abscess, pneumococcal pneumonia, and urinary tract infections). Methods Two-sample Mendelian randomization (MR) analysis was performed using inverse variance weighted (IVW), maximum likelihood, MR-Egger, weighted median, and MR-PRESSO. Results After adding other MR models and sensitivity analyses, genus Roseburia was simultaneously associated adversely with CRP (Beta IVW = -0.040) and SAA1 (Beta IVW = -0.280), and family Bifidobacteriaceae was negatively associated with both CRP (Beta IVW = -0.034) and pneumonia risk (Beta IVW = -0.391). After correction by FDR, only glutaroyl carnitine remained significantly associated with elevated CRP levels (Beta IVW = 0.112). Additionally, threonine (Beta IVW = 0.200) and 1-heptadecanoylglycerophosphocholine (Beta IVW = -0.246) were found to be significantly associated with WBC levels. Three metabolites showed similar causal effects on different inflammatory markers or infectious phenotypes, stearidonate (18:4n3) was negatively related to SAA1 and urinary tract infections, and 5-oxoproline contributed to elevated IL-6 and SAA1 levels. In addition, 7-methylguanine showed a positive correlation with dysentery and bacterial pneumonia. Conclusion This study provides novel evidence confirming the causal effects of the gut microbiome and the plasma metabolite profile on inflammation and the risk of infection. These potential molecular alterations may aid in the development of new targets for the intervention and management of disorders associated with inflammation and infections.
Collapse
Affiliation(s)
- Yingjian Liu
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Zhu
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong, China
| | - Gongjie Guo
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhipeng Xie
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Senlin Li
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chengyang Lai
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yonglin Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Liansheng Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shilong Zhong
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Vita AA, Roberts KM, Gundersen A, Farris Y, Zwickey H, Bradley R, Weir TL. Relationships between Habitual Polyphenol Consumption and Gut Microbiota in the INCLD Health Cohort. Nutrients 2024; 16:773. [PMID: 38542685 PMCID: PMC10974568 DOI: 10.3390/nu16060773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/01/2024] Open
Abstract
While polyphenol consumption is often associated with an increased abundance of beneficial microbes and decreased opportunistic pathogens, these relationships are not completely described for polyphenols consumed via habitual diet, including culinary herb and spice consumption. This analysis of the International Cohort on Lifestyle Determinants of Health (INCLD Health) cohort uses a dietary questionnaire and 16s microbiome data to examine relationships between habitual polyphenol consumption and gut microbiota in healthy adults (n = 96). In this exploratory analysis, microbial taxa, but not diversity measures, differed by levels of dietary polyphenol consumption. Taxa identified as exploratory biomarkers of daily polyphenol consumption (mg/day) included Lactobacillus, Bacteroides, Enterococcus, Eubacterium ventriosum group, Ruminococcus torques group, and Sutterella. Taxa identified as exploratory biomarkers of the frequency of polyphenol-weighted herb and spice use included Lachnospiraceae UCG-001, Lachnospiraceae UCG-004, Methanobrevibacter, Lachnoclostridium, and Lachnotalea. Several of the differentiating taxa carry out activities important for human health, although out of these taxa, those with previously described pro-inflammatory qualities in certain contexts displayed inverse relationships with polyphenol consumption. Our results suggest that higher quantities of habitual polyphenol consumption may support an intestinal environment where opportunistic and pro-inflammatory bacteria are represented in a lower relative abundance compared to those with less potentially virulent qualities.
Collapse
Affiliation(s)
- Alexandra Adorno Vita
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| | - Kristen M. Roberts
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, OH 43210, USA
| | - Anders Gundersen
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
| | - Yuliya Farris
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA 99352, USA
| | - Heather Zwickey
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
| | - Ryan Bradley
- Helfgott Research Institute, National University of Natural Medicine, Portland, OR 97201, USA
- Herbert Wertheim School of Public Health, University of California, San Diego, CA 92037, USA
| | - Tiffany L. Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
48
|
Langford L, Shah DD. Bioinformatic Analysis of Sulfotransferases from an Unexplored Gut Microbe, Sutterella wadsworthensis 3_1_45B: Possible Roles towards Detoxification via Sulfonation by Members of the Human Gut Microbiome. Int J Mol Sci 2024; 25:2983. [PMID: 38474230 DOI: 10.3390/ijms25052983] [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: 01/19/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Sulfonation, primarily facilitated by sulfotransferases, plays a crucial role in the detoxification pathways of endogenous substances and xenobiotics, promoting metabolism and elimination. Traditionally, this bioconversion has been attributed to a family of human cytosolic sulfotransferases (hSULTs) known for their high sequence similarity and dependence on 3'-phosphoadenosine 5'-phosphosulfate (PAPS) as a sulfo donor. However, recent studies have revealed the presence of PAPS-dependent sulfotransferases within gut commensals, indicating that the gut microbiome may harbor a diverse array of sulfotransferase enzymes and contribute to detoxification processes via sulfation. In this study, we investigated the prevalence of sulfotransferases in members of the human gut microbiome. Interestingly, we stumbled upon PAPS-independent sulfotransferases, known as aryl-sulfate sulfotransferases (ASSTs). Our bioinformatics analyses revealed that members of the gut microbial genus Sutterella harbor multiple asst genes, possibly encoding multiple ASST enzymes within its members. Fluctuations in the microbes of the genus Sutterella have been associated with various health conditions. For this reason, we characterized 17 different ASSTs from Sutterella wadsworthensis 3_1_45B. Our findings reveal that SwASSTs share similarities with E. coli ASST but also exhibit significant structural variations and sequence diversity. These differences might drive potential functional diversification and likely reflect an evolutionary divergence from their PAPS-dependent counterparts.
Collapse
Affiliation(s)
- Lauryn Langford
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| | - Dhara D Shah
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ 85281, USA
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA
| |
Collapse
|
49
|
Peng J, Cai K, Chen G, Liu L, Peng L. Genetic evidence strengthens the bidirectional connection between gut microbiota and Shigella infection: insights from a two-sample Mendelian randomization study. Front Microbiol 2024; 15:1361927. [PMID: 38495509 PMCID: PMC10941758 DOI: 10.3389/fmicb.2024.1361927] [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: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
Background In recent investigations, substantial strides have been made in the precise modulation of the gut microbiota to prevent and treat a myriad of diseases. Simultaneously, the pressing issue of widespread antibiotic resistance and multidrug resistance resulting from Shigella infections demands urgent attention. Several studies suggest that the antagonistic influence of the gut microbiota could serve as a novel avenue for impeding the colonization of pathogenic microorganisms or treating Shigella infections. However, conventional research methodologies encounter inherent challenges in identifying antagonistic microbial agents against Shigella, necessitating a comprehensive and in-depth analysis of the causal relationship between Shigella infections and the gut microbiota. Materials and methods Utilizing the aggregated summary statistics from Genome-Wide Association Studies (GWAS), we conducted Mendelian Randomization (MR) analyses encompassing 18,340 participants to explore the interplay between the gut microbiota and Shigella infections. This investigation also involved 83 cases of Shigella infection patients and 336,396 control subjects. In the positive strand of our findings, we initially performed a preliminary analysis using the Inverse Variance Weighting (IVW) method. Subsequently, we undertook sensitivity analyses to assess the robustness of the results, addressing confounding factors' influence. This involved employing the Leave-One-Out method and scrutinizing funnel plots to ensure the reliability of the MR analysis outcomes. Conclusively, a reverse MR analysis was carried out, employing the Wald ratio method due to the exposure of individual Single Nucleotide Polymorphisms (SNPs). This was undertaken to explore the plausible associations between Shigella infections and genetically predicted compositions of the gut microbiota. Results In this study, we employed 2,818 SNPs associated with 211 species of gut microbiota as instrumental variables (IVs). Through IVW analysis, our positive MR findings revealed a significant negative correlation between the occurrence of Shigella infections and the phylum Tenericutes (OR: 0.18, 95% CI: 0.04-0.74, p = 0.02), class Mollicutes (OR: 0.18, 95% CI: 0.04-0.74, p = 0.02), genus Intestinimonas (OR: 0.16, 95% CI: 0.04-0.63, p = 0.01), genus Gordonibacter (OR: 0.39, 95% CI: 0.16-0.93, p = 0.03), and genus Butyrivibrio (OR: 0.44, 95% CI: 0.23-0.87, p = 0.02). Conversely, a positive correlation was observed between the occurrence of Shigella infections and genus Sutterella (OR: 10.16, 95% CI: 1.87-55.13, p = 0.01) and genus Alistipes (OR: 12.24, 95% CI: 1.71-87.34, p = 0.01). In sensitivity analyses, utilizing MR-Egger regression analysis and MR Pleiotropy Residual Sum and Outlier (MR-PRESSO) detection, all outcomes demonstrated robust stability. Simultaneously, in the reverse MR analysis, Shigella infections resulted in an upregulation of four bacterial genera and a downregulation of three bacterial genera. Conclusion In summation, the MR analysis outcomes corroborate the presence of bidirectional causal relationships between the gut microbiota and Shigella infections. This study not only unveils novel perspectives for the prevention and treatment of Shigella infections but also furnishes fresh insights into the mechanistic underpinnings of how the gut microbiota contributes to the pathogenesis of Shigella infections. Consequently, the established dual causal association holds promise for advancing our understanding and addressing the complexities inherent in the interplay between the gut microbiota and Shigella infections, thereby paving the way for innovative therapeutic interventions and preventive strategies in the realm of Shigella-related diseases.
Collapse
Affiliation(s)
- Jingyi Peng
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Kun Cai
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Linxiao Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Lili Peng
- The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, China
| |
Collapse
|
50
|
Gilley SP, Zarate MA, Zheng L, Jambal P, Yazza DN, Chintapalli SV, MacLean PS, Wright CJ, Rozance PJ, Shankar K. Metabolic and fecal microbial changes in adult fetal growth restricted mice. Pediatr Res 2024; 95:647-659. [PMID: 37935884 PMCID: PMC10899111 DOI: 10.1038/s41390-023-02869-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Fetal growth restriction (FGR) increases risk for development of obesity and type 2 diabetes. Using a mouse model of FGR, we tested whether metabolic outcomes were exacerbated by high-fat diet challenge or associated with fecal microbial taxa. METHODS FGR was induced by maternal calorie restriction from gestation day 9 to 19. Control and FGR offspring were weaned to control (CON) or 45% fat diet (HFD). At age 16 weeks, offspring underwent intraperitoneal glucose tolerance testing, quantitative MRI body composition assessment, and energy balance studies. Total microbial DNA was used for amplification of the V4 variable region of the 16 S rRNA gene. Multivariable associations between groups and genera abundance were assessed using MaAsLin2. RESULTS Adult male FGR mice fed HFD gained weight faster and had impaired glucose tolerance compared to control HFD males, without differences among females. Irrespective of weaning diet, adult FGR males had depletion of Akkermansia, a mucin-residing genus known to be associated with weight gain and glucose handling. FGR females had diminished Bifidobacterium. Metabolic changes in FGR offspring were associated with persistent gut microbial changes. CONCLUSION FGR results in persistent gut microbial dysbiosis that may be a therapeutic target to improve metabolic outcomes. IMPACT Fetal growth restriction increases risk for metabolic syndrome later in life, especially if followed by rapid postnatal weight gain. We report that a high fat diet impacts weight and glucose handling in a mouse model of fetal growth restriction in a sexually dimorphic manner. Adult growth-restricted offspring had persistent changes in fecal microbial taxa known to be associated with weight, glucose homeostasis, and bile acid metabolism, particularly Akkermansia, Bilophilia and Bifidobacteria. The gut microbiome may represent a therapeutic target to improve long-term metabolic outcomes related to fetal growth restriction.
Collapse
Affiliation(s)
- Stephanie P Gilley
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Miguel A Zarate
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Purevsuren Jambal
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deaunabah N Yazza
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Paul S MacLean
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Clyde J Wright
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paul J Rozance
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kartik Shankar
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|