1
|
Pang F, Jiang Q, Li K, Tang X. Integrative gut microbiota, metabolomics and proteomics studies unraveled the mechanism of Shaoteng decoction in treating Sjogren's syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156631. [PMID: 40088738 DOI: 10.1016/j.phymed.2025.156631] [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: 12/03/2024] [Revised: 02/17/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Sjögren's syndrome (SS) is a complicated autoimmune disorder, encompassing multifaceted pathogenesis of inflammatory response, immune dysregulation and metabolic abnormalities. Shaoteng Decoction (STD) is a type of traditional Chinese medicine preparation that has been shown to effectively improve inflammatory damage and immune dysfunction in patients with SS. Nevertheless, the exact mechanism has not been unspecified. PURPOSE This work aims to determine the mechanism of STD treatment on SS, identifying potential therapeutic targets and their relationships. METHODS Non-obese diabetic mice served as a disease model. This study analyzes potential signaling pathways of STD treatment for SS through network pharmacology, and assesses the role of STD in reducing inflammatory damage using pathological staining, ELISA, and immunohistochemistry. Additionally, the study apply gut microbiota, metabolomics, and proteomics analyses to identify the key microbiota, metabolites and proteins, aiming to find potential action targets of STD. We use Western blotting and immunohistochemistry to verify the authenticity of the relevant targets and study the interactions among gut microbiota, metabolites, and proteins. RESULTS Proteobacteria is the important intestinal bacteria, Bile Acid Biosynthesis is the main metabolic pathway, IfI30, Ndufv3, and Ndufs6 are the crucial differential expressed proteins. Moreover, there is a strong correlation among the three. STD treats SS by reducing the abundance of Proteobacteria, increasing Bile Acid Biosynthesis, decreasing IfI30 expression, and increasing the expression of Ndufv3 and Ndufs6. CONCLUSION STD inhibits inflammatory responses, improves immune dysregulation and energy metabolism abnormalities, and prevents the progression of SS through regulating the gut microbiota, enhancing Bile Acid Biosynthesis, and modulating proteins expression levels.
Collapse
Affiliation(s)
- Fengtao Pang
- Department of Rheumatology, Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Quan Jiang
- Department of Rheumatology, Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Kesong Li
- Department of Rheumatology, Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, People's Republic of China.
| | - Xiaopo Tang
- Department of Rheumatology, Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, People's Republic of China.
| |
Collapse
|
2
|
Pasha A, Iqbal NT, Shafiq Y, Khan W, Azam SI, Kabir F, Muhammad A, Nisar MI, Jehan F. Effect of one prophylactic dose of azithromycin on Bifidobacterium infantis colonization in infants from the Mumta trial. Int J Infect Dis 2025; 153:107794. [PMID: 39855335 PMCID: PMC11910343 DOI: 10.1016/j.ijid.2025.107794] [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: 10/31/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVES The effects of antibiotics on the microbiome remain incompletely understood. Azithromycin (AZ) has been shown to improve child survival and infant growth outcomes. This study aimed to assess the impact of AZ on Bifidobacterium infantis colonization and bacterial enteropathogen count in the infant gut. METHODS We analyzed clinical, biomarker, B. infantis and enteropathogen data from 150 mother-infant dyads from the MUMTA Lactating Women study. Colonization of B. infantis was assessed using quantitative polymerase chain reaction (PCR) of fecal samples. We utilized a customized PCR-based TaqMan Array Card for enteropathogen detection. RESULTS AZ administration was associated with a 1.99-fold (95% confidence intervals [CI] 1.33-2.97) increase in colonization by B. infantis. B. infantis colonization was highest when inflammatory biomarker levels were within normal range. Mode of delivery (RR 2.43; 95% CI: 1.58, 3.76) and colostrum (RR2.05; 95% CI: 1.41, 2.98) given to the infant within 24 h of birth were associated with B. infantis colonization. A single dose of AZ on day 42 reduced bacterial enteropathogen count in the AZ group on day 56, as compared to the pre-AZ count. Bacterial enteropathogen count for infants with wasting (weight for length z-score WLZ <-2) was 1.43-fold higher (95% CI: 1.00-2.03) than for infants with WLZ ≥ -2. Over 60% of infants harbored with the macrolide resistance mph(A) gene CONCLUSION: AZ administration increases B. infantis colonization and reduces bacterial enteropathogen count in infants.
Collapse
Affiliation(s)
- Aneela Pasha
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Najeeha Talat Iqbal
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan; Department of Biological & Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Yasir Shafiq
- Center of Excellence for Trauma and Emergencies and Community Health Sciences, The Aga Khan University, Karachi, Pakistan; Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Waqasuddin Khan
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Syed Iqbal Azam
- Department of Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | | | - Muhammad Imran Nisar
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Fyezah Jehan
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
3
|
Azpiazu-Muniozguren M, García-Martínez M, Zabaleta A, Antiguedad I, Garaizar J, Laorden L, Martinez-Malaxetxebarria I, Martinez-Ballesteros I. Prokaryotic Diversity and Community Distribution in the Complex Hydrogeological System of the Añana Continental Saltern. MICROBIAL ECOLOGY 2025; 87:171. [PMID: 39820572 PMCID: PMC11739210 DOI: 10.1007/s00248-025-02488-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Abstract
The Añana Salt Valley (northern Spain) is a continental saltern consisting of a series of natural springs that have been used for salt production for at least 7000 years. This habitat has been relatively understudied; therefore, prokaryotic diversity was investigated through Illumina-based 16S rRNA gene sequencing to determine if the waters within the valley exhibit distinctive microbiological characteristics. Two main types of water were found in the valley: salty (approximately 200 g/L salinity) from the diapiric structure and brackish (≤ 20 g/L salinity) from shallow streams. The beta diversity indices showed that salinity was the primary factor influencing the prokaryotic distribution. However, a niche-specific influence was observed between waters of the same origin, with significant differences in the relative abundance of the ASVs. The microbiome of the saltern revealed that the archaeal domain was mainly restricted to salty waters, while the bacterial domain was ubiquitous throughout the saltern, with a notable prevalence in brackish waters. The main bacterial and archaeal phyla identified were Pseudomonadota and Halobacterota, respectively. The genus Halorubrum was abundant and widespread in salty waters, while Pseudomonas was a significant part of the prokaryote community, mainly in brackish waters. The relative abundance of the genera Haloplanus and Salinibacter increased in the salt ponds used for salt production. The taxa involved in chemoheterotrophy and fermentation were widespread, sharing the same niche. Overall, the location of this saltern on a diapiric structure favors the occurrence of waters with different origins that affect the prokaryotic distribution beyond the niche location in the valley.
Collapse
Affiliation(s)
- Maia Azpiazu-Muniozguren
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Minerva García-Martínez
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Ane Zabaleta
- Hydro-Environment Processes Research Group. Geology Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, Barrio Sarriena S/N, 48940, Leioa, Spain
| | - Iñaki Antiguedad
- Hydro-Environment Processes Research Group. Geology Department, Faculty of Science and Technology, University of the Basque Country UPV/EHU, Barrio Sarriena S/N, 48940, Leioa, Spain
| | - Javier Garaizar
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Diseases, Antimicrobial Agents, and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Lorena Laorden
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Diseases, Antimicrobial Agents, and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Irati Martinez-Malaxetxebarria
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Microbiology, Infectious Diseases, Antimicrobial Agents, and Gene Therapy, 01006, Vitoria-Gasteiz, Spain
| | - Ilargi Martinez-Ballesteros
- MikroIker Research Group, Immunology, Microbiology and Parasitology Department, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de La Universidad 7, 01006, Vitoria-Gasteiz, Spain.
- Bioaraba, Microbiology, Infectious Diseases, Antimicrobial Agents, and Gene Therapy, 01006, Vitoria-Gasteiz, Spain.
| |
Collapse
|
4
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Sabaredzovic A, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Philippat C, Eggesbø M, Lepage P, Slama R. Associations between pre- and post-natal exposure to phthalate and DINCH metabolites and gut microbiota in one-year old children. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125204. [PMID: 39490662 DOI: 10.1016/j.envpol.2024.125204] [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: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The gut microbiota is a collection of symbiotic microorganisms in the gastrointestinal tract. Its sensitivity to chemicals with widespread exposure, such as phthalates, is little known. We aimed to investigate the impact of perinatal exposure to phthalates on the infant gut microbiota at 12 months of age. Within SEPAGES cohort (Suivi de l'Exposition à la Pollution Atmosphérique durant la Grossesse et Effet sur la Santé), we assessed 13 phthalate metabolites and 2 di(isononyl) cyclohexane-1,2-dicarboxylate (DINCH) metabolites in repeated urine samples collected in pregnant women and their offspring. We obtained stool samples from 356 children at 12 months of age and sequenced the V3-V4 region of the 16S rRNA gene, allowing gut bacterial profiling. We used single-chemical (linear regressions) and mixture (BKMR, Bayesian Kernel Machine Regression) models to examine associations of phthalates and DINCH metabolites, with gut microbiota indices of α-diversity (specific richness and Shannon diversity) and the relative abundances of the most abundant microbiota phyla and genera. After correction for multiple testing, di(2-ethylhexyl) phthalate (ΣDEHP), diethyl phthalate (DEP) and bis(2-propylheptyl) phthalate (DPHP) metabolites 12-month urinary concentrations were associated with higher Shannon α-diversity of the child gut microbiota in single-chemical models. The multiple-chemical model (BKMR) suggested higher α-diversity with exposure to the phthalate mixture at 12 months, driven by the same phthalates. There were no associations between phthalate and DINCH exposure biomarkers at other time points and α-diversity after correction for multiple testing. ΣDEHP metabolites concentration at 12 months was associated with higher Coprococcus genus. Finally, ΣDEHP exposure at 12 months tended to be associated with higher phylum Firmicutes, an association not maintained after correction for multiple testing. Infancy exposure to phthalate might disrupt children's gut microbiota. The observed associations were cross-sectional, so that reverse causality cannot be excluded.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France.
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Azemira Sabaredzovic
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), 0213, Oslo, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France
| | - Merete Eggesbø
- Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway; Faculty of Veterinary Medicine, Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, F-78350, Jouy-en-Josas, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, University Grenoble Alpes, 38000, Grenoble, France; SMILE, Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France; PARSEC, Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005, Paris, France
| |
Collapse
|
5
|
Davias A, Lyon-Caen S, Rolland M, Iszatt N, Thomsen C, Haug LS, Sakhi AK, Monot C, Rayah Y, Ilhan ZE, Jovanovic N, Philippat C, Eggesbo M, Lepage P, Slama R. Perinatal Exposure to Phenols and Poly- and Perfluoroalkyl Substances and Gut Microbiota in One-Year-Old Children. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15395-15414. [PMID: 39173114 DOI: 10.1021/acs.est.3c09927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The role of the gut microbiota in human health calls for a better understanding of its determinants. In particular, the possible effects of chemicals with widespread exposure other than pharmaceuticals are little known. Our aim was to characterize the sensitivity of the early-life gut microbiota to specific chemicals with possible antimicrobial action. Within the SEPAGES French couple-child cohort, we assessed 12 phenols in repeated urine samples from 356 pregnant women and their offspring and 19 poly- and perfluoroalkyl substances (PFASs) in serum from the pregnant women. We collected stool samples from the children at one year of age, in which the V3-V4 region of the 16S rRNA gene was sequenced, allowing for gut bacterial profiling. Associations of each chemical with α- and β-diversity indices of the gut microbiota and with the relative abundance of the most abundant taxa were assessed using single-pollutant and mixture (BKMR) models. Perinatal exposure to certain parabens was associated with gut microbiota α- and β-diversity and with Firmicutes and Proteobacteria. Suggestive associations of certain phenols with genera of the Lachnospiraceae and Enterobacteriaceae families were observed, but these were not maintained after correction for multiple testing. Parabens, which have known antimicrobial properties, might disrupt the child gut microbiota, but larger studies are required to confirm these findings.
Collapse
Affiliation(s)
- Aline Davias
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Sarah Lyon-Caen
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Matthieu Rolland
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Nina Iszatt
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Cathrine Thomsen
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Line Småstuen Haug
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Amrit Kaur Sakhi
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Celine Monot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Yamina Rayah
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Zehra Esra Ilhan
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Nicolas Jovanovic
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Claire Philippat
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| | - Merete Eggesbo
- Division of Climate and Environmental Health, Norwegian Institute of Public Health (NIPH), Oslo 0213, Norway
| | - Patricia Lepage
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Rémy Slama
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, La Tronche 38700, France
| |
Collapse
|
6
|
Ezenabor EH, Adeyemi AA, Adeyemi OS. Gut Microbiota and Metabolic Syndrome: Relationships and Opportunities for New Therapeutic Strategies. SCIENTIFICA 2024; 2024:4222083. [PMID: 39041052 PMCID: PMC11262881 DOI: 10.1155/2024/4222083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/10/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Since its discovery, numerous studies have shown the role of the microbiota in well-being and disease. The gut microbiota represents an essential factor that plays a multidirectional role that affects not just the gut but also other parts of the body, including the brain, endocrine system, humoral system, immune system, and metabolic pathways, as well as host-microbiome interactions. Through a comprehensive analysis of existing literature using the desktop research methodology, this review elucidates the mechanisms by which gut microbiota dysbiosis contributes to metabolic dysfunction, including obesity, dyslipidaemia, hypertension, atherosclerosis, hyperuricemia, and hyperglycaemia. Furthermore, it examines the bidirectional communication pathways between gut microbiota and host metabolism, highlighting the role of microbial-derived metabolites, immune modulation, and gut barrier integrity in shaping metabolic homeostasis. Importantly, the review identifies promising therapeutic strategies targeting the gut microbiota as potential interventions for metabolic syndrome, including probiotics, prebiotics, symbiotics, dietary modifications, and faecal microbiota transplantation. By delineating the bidirectional interactions between gut microbiota and metabolic syndrome, the review not only advances our understanding of disease pathophysiology but also underscores the potential for innovative microbiota-based interventions to mitigate the global burden of metabolic syndrome and its associated complications.
Collapse
Affiliation(s)
- Emmanuel Henry Ezenabor
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Aishat Abimbola Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Oluyomi Stephen Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| |
Collapse
|
7
|
Qadri H, Shah AH, Almilaibary A, Mir MA. Microbiota, natural products, and human health: exploring interactions for therapeutic insights. Front Cell Infect Microbiol 2024; 14:1371312. [PMID: 39035357 PMCID: PMC11257994 DOI: 10.3389/fcimb.2024.1371312] [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: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/23/2024] Open
Abstract
The symbiotic relationship between the human digestive system and its intricate microbiota is a captivating field of study that continues to unfold. Comprising predominantly anaerobic bacteria, this complex microbial ecosystem, teeming with trillions of organisms, plays a crucial role in various physiological processes. Beyond its primary function in breaking down indigestible dietary components, this microbial community significantly influences immune system modulation, central nervous system function, and disease prevention. Despite the strides made in microbiome research, the precise mechanisms underlying how bacterial effector functions impact mammalian and microbiome physiology remain elusive. Unlike the traditional DNA-RNA-protein paradigm, bacteria often communicate through small molecules, underscoring the imperative to identify compounds produced by human-associated bacteria. The gut microbiome emerges as a linchpin in the transformation of natural products, generating metabolites with distinct physiological functions. Unraveling these microbial transformations holds the key to understanding the pharmacological activities and metabolic mechanisms of natural products. Notably, the potential to leverage gut microorganisms for large-scale synthesis of bioactive compounds remains an underexplored frontier with promising implications. This review serves as a synthesis of current knowledge, shedding light on the dynamic interplay between natural products, bacteria, and human health. In doing so, it contributes to our evolving comprehension of microbiome dynamics, opening avenues for innovative applications in medicine and therapeutics. As we delve deeper into this intricate web of interactions, the prospect of harnessing the power of the gut microbiome for transformative medical interventions becomes increasingly tantalizing.
Collapse
Affiliation(s)
- Hafsa Qadri
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Abdul Haseeb Shah
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Abdullah Almilaibary
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Family and Community Medicine, Faculty of Medicine, Al Baha University, Al Bahah, Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| |
Collapse
|
8
|
Vohra A, Karnik R, Desai M, Vyas H, Kulshrestha S, Upadhyay KK, Koringa P, Devkar R. Melatonin-mediated corrective changes in gut microbiota of experimentally chronodisrupted C57BL/6J mice. Chronobiol Int 2024; 41:548-560. [PMID: 38557404 DOI: 10.1080/07420528.2024.2329205] [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: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Chronic consumption of a high-calorie diet coupled with an altered sleep-wake cycle causes disruption of circadian clock that can impact the gut microbiome leading to metabolic syndrome and associated diseases. Herein, we investigate the effects of a high fat high fructose diet (H) alone or in combination with photoperiodic shifts induced chronodisruption (CD) on gut microbiota of C57BL/6J male mice. Further, the merits of daily evening intraperitoneal administration of melatonin in restoring gut microbiota are studied herein. Experimental groups viz. H, CD and HCD mice recorded higher levels of serum pro-inflammatory cytokines (TNF-α and IL-6) and lower levels of the anti-inflammatory cytokine, IL-10. These findings correlate with a concomitant increase in the transcripts of TLR4, TNF-α, and IL-6 in small intestine of the said groups. A decrement in mRNA levels of Ocln, ZO-1 and Vdr in these groups implied towards an altered gut permeability. These results were in agreement with the observed decrement in percentage abundance of total gut microflora and Firmicutes: Bacteroidetes (F/B) ratio. Melatonin administration accounted for lower-level inflammation (serum and gut) along with an improvement in gut permeability markers. The total abundance of gut microflora and F/B ratio showed an improvement in all the melatonin-treated groups and the same is the highlight of this study. Taken together, our study is the first to report perturbations in gut microbiota resulting due to a combination of photoperiodic shifts induced CD and a high fat high calorie diet-induced lifestyle disorder. Further, melatonin-mediated rejuvenation of gut microbiome provides prima facie evidence of its role in improving gut dysbiosis that needs a detailed scrutiny.
Collapse
Affiliation(s)
- Aliasgar Vohra
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Department of Neurology, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Rhydham Karnik
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
- Dr Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mansi Desai
- Department of Animal Biotechnology, College of Veterinary Sciences & A.H., Anand Agricultural University, Anand, India
| | - Hitarthi Vyas
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shruti Kulshrestha
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Kapil Kumar Upadhyay
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Prakash Koringa
- Department of Animal Biotechnology, College of Veterinary Sciences & A.H., Anand Agricultural University, Anand, India
| | - Ranjitsinh Devkar
- Division of Chronobiology and Metabolic Endocrinology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
9
|
Sundararaman A, Halami PM. Metabolic Engineering of Bifidobacterium sp. Using Genome Editing Techniques. GENOME EDITING IN BACTERIA (PART 1) 2024:88-105. [DOI: 10.2174/9789815165678124010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The gut microbiome is significant in maintaining human health by
facilitating absorption and digestion in the intestine. Probiotics have diverse and
significant applications in the health sector, so probiotic strains require an
understanding of the genome-level organizations. Probiotics elucidate various
functional parameters that control their metabolic functions. Gut dysbiosis leads to
inflammatory bowel disease and other neurological disorders. The application of
probiotic bacteria to modulate the gut microbiota prevents diseases and has gained
large interest. In a recent decade, the development of modern tools in molecular
biology has led to the discovery of genome engineering. Synthetic biology approaches
provide information about diverse biosynthetic pathways and also facilitate novel
metabolic engineering approaches for probiotic strain improvement. The techniques
enable engineering probiotics with the desired functionalities to benefit human health.
This chapter describes the recent advances in probiotic strain improvement for
diagnostic and therapeutic applications via CRISPR-Cas tools. Also, the application of
probiotics, current challenges, and future perspectives in disease treatment are
discussed.
Collapse
Affiliation(s)
- Aravind Sundararaman
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological
Research Institute, Mysuru-570020, India
| | - Prakash M. Halami
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological
Research Institute, Mysuru-570020, India
| |
Collapse
|
10
|
Feng P, Xue X, Bukhari I, Qiu C, Li Y, Zheng P, Mi Y. Gut microbiota and its therapeutic implications in tumor microenvironment interactions. Front Microbiol 2024; 15:1287077. [PMID: 38322318 PMCID: PMC10844568 DOI: 10.3389/fmicb.2024.1287077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its tumor microenvironment (TME) also coevolves with it, which is primarily involved in tumor initiation, development, metastasis, and therapeutic responses. Recent years, TME has been emerged as a potential target for cancer diagnosis and treatment. However, the clinical efficacy of treatments targeting the TME, especially its specific components, remains insufficient. In parallel, the gut microbiome is an essential TME component that is crucial in cancer immunotherapy. Thus, assessing and constructing frameworks between the gut microbiota and the TME can significantly enhance the exploration of effective treatment strategies for various tumors. In this review the role of the gut microbiota in human cancers, including its function and relationship with various tumors was summarized. In addition, the interaction between the gut microbiota and the TME as well as its potential applications in cancer therapeutics was described. Furthermore, it was summarized that fecal microbiota transplantation, dietary adjustments, and synthetic biology to introduce gut microbiota-based medical technologies for cancer treatment. This review provides a comprehensive summary for uncovering the mechanism underlying the effects of the gut microbiota on the TME and lays a foundation for the development of personalized medicine in further studies.
Collapse
Affiliation(s)
- Pengya Feng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Children Rehabilitation Medicine, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xia Xue
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ihtisham Bukhari
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunjing Qiu
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Li
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Sharma D, Gajjar D, Seshadri S. Understanding the role of gut microfloral bifidobacterium in cancer and its potential therapeutic applications. MICROBIOME RESEARCH REPORTS 2023; 3:3. [PMID: 38455077 PMCID: PMC10917622 DOI: 10.20517/mrr.2023.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 03/09/2024]
Abstract
Gut microbiota research has gained a tremendous amount of attention from the scientific community because of its contribution to gut homeostasis, human health, and various pathophysiological conditions. The early colonizer of the human gut, i.e., bifidobacteria, has emerged as an efficient probiotic in various diseased conditions, including cancer. This review explores the pros and cons of Bifidobacterium in various malignancies and various therapeutic strategies. We have illustrated the controversial role of bifidobacteria participating in various malignancies as well as described the current knowledge regarding its use in anticancer therapies. Ultimately, this article also addresses the need for further extensive research in elucidating the mechanism of how bifidobacteria is involved and is indirectly affecting the tumor microenvironment. Exhaustive and large-scale research is also required to solve the controversial questions regarding the involvement of bifidobacteria in cancer research.
Collapse
Affiliation(s)
| | | | - Sriram Seshadri
- Institute of Science, Nirma University, 382481 Ahmedabad, Gujarat, India
| |
Collapse
|
12
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
13
|
Nishio M, Morioka H, Takai S, Osada Y, Seki Y, Osugi T, Oba A, Miyaki Y. Bacteraemia and obstructive pyelonephritis caused by Bifidobacterium breve in an elderly woman: a case report and literature review. Access Microbiol 2023; 5:000574.v3. [PMID: 37970080 PMCID: PMC10634491 DOI: 10.1099/acmi.0.000574.v3] [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: 02/01/2023] [Accepted: 07/19/2023] [Indexed: 11/17/2023] Open
Abstract
Bifidobacterium spp. are non-spore-forming Gram-positive anaerobes that are indigenous to the human gastrointestinal tract and vagina. They are believed to be non-pathogenic organisms for humans and thus are widely used as probiotics. An 83-year-old woman taking cephalexin for 4 days was diagnosed with obstructive pyelonephritis. Y-branched Gram-positive rods were found in both anaerobic and aerobic blood culture bottles, and in an anaerobic urine culture. Bifidobacterium breve was finally identified. Ceftriaxone and metronidazole were administered to the patient, and she was discharged after intermittent catheterization for dysuria. Urinary tract infection caused by Bifidobacterium spp. is believed to be rare, but it can develop in patients with underlying urological conditions. Recognition of the characteristic morphology and conducting anaerobic urine culture may help in identifying more cases of Bifidobacterium urinary tract infections.
Collapse
Affiliation(s)
- Mitsuru Nishio
- Department of Clinical Laboratory, Komaki City Hospital, Komaki, Aichi, Japan
- Infection Control Team, Komaki City Hospital, Komaki, Aichi, Japan
| | - Hiroshi Morioka
- Infection Control Team, Komaki City Hospital, Komaki, Aichi, Japan
- Department of Infectious Diseases, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Shun Takai
- Department of Urology, Komaki City Hospital, Komaki, Aichi, Japan
| | - Yukari Osada
- Department of Medical Technique, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Yoshie Seki
- Department of Clinical Laboratory, Komaki City Hospital, Komaki, Aichi, Japan
| | - Takato Osugi
- Department of Clinical Laboratory, Komaki City Hospital, Komaki, Aichi, Japan
| | - Airi Oba
- Department of Clinical Laboratory, Komaki City Hospital, Komaki, Aichi, Japan
| | - Yuki Miyaki
- Department of Clinical Laboratory, Komaki City Hospital, Komaki, Aichi, Japan
| |
Collapse
|
14
|
Najafpour B, Pinto PIS, Sanz EC, Martinez-Blanch JF, Canario AVM, Moutou KA, Power DM. Core microbiome profiles and their modification by environmental, biological, and rearing factors in aquaculture hatcheries. MARINE POLLUTION BULLETIN 2023; 193:115218. [PMID: 37441915 DOI: 10.1016/j.marpolbul.2023.115218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/06/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023]
Abstract
16S rRNA gene sequencing and bacteria- and genus-specific quantitative PCR was used to profile microbial communities and their associated functions in water, live feed (microalgae, Artemia, and rotifer), and European sea bass and gilthead sea bream larvae from hatcheries in Greece and Italy. The transfer to larvae of genus containing potential pathogens of fish was more likely with Artemia and rotifer than with microalgae or water, irrespective of geographic location. The presence of potentially pathogenic bacteria (Vibrio and Pseudoalteromonas) in the core microbiota of water, live feed, and fish larvae, the enrichment of different bacterial resistance pathways and biofilm formation, and the overall low beneficial bacteria load during larval ontogeny emphasizes the risk for disease outbreaks. The present data characterizing microbiota in commercial aquaculture hatcheries provides a baseline for the design of strategies to manage disease and to model or remediate potential adverse environmental impacts.
Collapse
Affiliation(s)
- Babak Najafpour
- CCMAR - Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, Edifício 7, 8005-139 Faro, Portugal
| | - Patricia I S Pinto
- CCMAR - Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, Edifício 7, 8005-139 Faro, Portugal
| | - Eric Climent Sanz
- ADM Biopolis, Parc Cientific Universidad De Valencia, Paterna, Spain
| | | | - Adelino V M Canario
- CCMAR - Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, Edifício 7, 8005-139 Faro, Portugal; Shanghai Ocean University International Center for Marine Studies, Shanghai, China
| | - Katerina A Moutou
- Department of Biochemistry & Biotechnology, University of Thessaly, Ploutonos 26, Larissa, Greece
| | - Deborah M Power
- CCMAR - Centre of Marine Sciences, Universidade do Algarve, Campus de Gambelas, Edifício 7, 8005-139 Faro, Portugal; Shanghai Ocean University International Center for Marine Studies, Shanghai, China.
| |
Collapse
|
15
|
Nishiyama K, Yong CC, Moritoki N, Kitazawa H, Odamaki T, Xiao JZ, Mukai T. Sharing of Moonlighting Proteins Mediates the Symbiotic Relationship among Intestinal Commensals. Appl Environ Microbiol 2023; 89:e0219022. [PMID: 36847513 PMCID: PMC10053696 DOI: 10.1128/aem.02190-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
The human gastrointestinal tract is inhabited by trillions of symbiotic bacteria that form a complex ecological community and influence human physiology. Symbiotic nutrient sharing and nutrient competition are the most studied relationships in gut commensals, whereas the interactions underlying homeostasis and community maintenance are not fully understood. Here, we provide insights into a new symbiotic relationship wherein the sharing of secreted cytoplasmic proteins, called "moonlighting proteins," between two heterologous bacterial strains (Bifidobacterium longum and Bacteroides thetaiotaomicron) was observed to affect the adhesion of bacteria to mucins. B. longum and B. thetaiotaomicron were cocultured using a membrane-filter system, and in this system the cocultured B. thetaiotaomicron cells showed greater adhesion to mucins compared to that shown by monoculture cells. Proteomic analysis showed the presence of 13 B. longum-derived cytoplasmic proteins on the surface of B. thetaiotaomicron. Moreover, incubation of B. thetaiotaomicron with the recombinant proteins GroEL and elongation factor Tu (EF-Tu)-two well-known mucin-adhesive moonlighting proteins of B. longum-led to an increase in the adhesion of B. thetaiotaomicron to mucins, a result attributed to the localization of these proteins on the B. thetaiotaomicron cell surface. Furthermore, the recombinant EF-Tu and GroEL proteins were observed to bind to the cell surface of several other bacterial species; however, the binding was species dependent. The present findings indicate a symbiotic relationship mediated by the sharing of moonlighting proteins among specific strains of B. longum and B. thetaiotaomicron. IMPORTANCE The adhesion of intestinal bacteria to the mucus layer is an important colonization strategy in the gut environment. Generally, the bacterial adhesion process is a characteristic feature of the individual cell surface-associated adhesion factors secreted by a particular bacterium. In this study, coculture experiments between Bifidobacterium and Bacteroides show that the secreted moonlighting proteins adhere to the cell surface of coexisting bacteria and alter the adhesiveness of the bacteria to mucins. This finding indicates that the moonlighting proteins act as adhesion factors for not only homologous strains but also for coexisting heterologous strains. The presence of a coexisting bacterium in the environment can significantly alter the mucin-adhesive properties of another bacterium. The findings from this study contribute to a better understanding of the colonization properties of gut bacteria through the discovery of a new symbiotic relationship between them.
Collapse
Affiliation(s)
- Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Cheng-Chung Yong
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Aobaku, Sendai, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Takao Mukai
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
16
|
Murugkar P, Dimise E, Stewart E, Viala SN, Clardy J, Dewhirst FE, Lewis K. Identification of a growth factor required for culturing specific fastidious oral bacteria. J Oral Microbiol 2023; 15:2143651. [DOI: 10.1080/20002297.2022.2143651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Pallavi Murugkar
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| | - Eric Dimise
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School 02115, Boston, MA, USA
| | - Eric Stewart
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| | - Stéphane N. Viala
- Department of Microbiology, the Forsyth Institute, Cambridge, MA, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School 02115, Boston, MA, USA
| | - Floyd E. Dewhirst
- Department of Microbiology, the Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| |
Collapse
|
17
|
Plaza-Díaz J, Manzano M, Ruiz-Ojeda FJ, Giron MD, Salto R, López-Pedrosa JM, Santos-Fandila A, Garcia-Corcoles MT, Rueda R, Gil Á. Intake of slow-digesting carbohydrates is related to changes in the microbiome and its functional pathways in growing rats with obesity induced by diet. Front Nutr 2022; 9:992682. [PMID: 36532542 PMCID: PMC9748084 DOI: 10.3389/fnut.2022.992682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/02/2022] [Indexed: 08/17/2023] Open
Abstract
INTRODUCTION The main cause of insulin resistance in childhood is obesity, which contributes to future comorbidities as in adults. Although high-calorie diets and lack of exercise contribute to metabolic disease development, food quality rather than the quantity of macronutrients is more important than food density. The purpose of the present study was to examine the effects of changing the quality of carbohydrates from rapidly to slowly digestible carbohydrates on the composition of the gut microbiota and the profiles of the functional pathways in growing rats with obesity due to a high-fat diet (HFD). METHODS During the course of 4 weeks, rats growing on an HFD-containing carbohydrates with different digestive rates were fed either HFD-containing carbohydrates with a rapid digestion rate (OBE group) or HFD-containing carbohydrates with a slow digestion rate (OBE-ISR group). A non-obese group (NOB) was included as a reference, and rats were fed on a rodent standard diet (AIN93G). An analysis of gut microbiota was conducted using 16S rRNA-based metagenomics; a linear mixed-effects model (LMM) was used to determine changes in abundance between baseline and 4 weeks of treatment, and functional pathways were identified. Gut microbiota composition at bacterial diversity and relative abundance, at phylum and genus levels, and functional profiles were analyzed by integrating the Integrated Microbial Genomes (IMG) database. RESULTS The groups showed comparable gut microbiota at baseline. At the end of the treatment, animals from the ISR group exhibited differences at the phylum levels by decreasing the diversity of Fisher's index and Firmicutes (newly named as Bacillota), and increasing the Pielou's evenness and Bacteroidetes (newly named as Bacteroidota); at the genus level by increasing Alistipes, Bifidobacterium, Bacteroides, Butyricimonas, Lachnoclostridium, Flavonifractor, Ruminiclostridium 5, and Faecalibaculum and decreasing Muribaculum, Blautia, and Ruminiclostridium 9. Remarkably, relative abundances of genera Tyzzerella and Angelakisella were higher in the OBE group compared to NOB and OBE-ISR groups. In addition, some microbiota carbohydrate metabolism pathways such as glycolysis, glucuronic acid degradation, pentose phosphate pathway, methanogenesis, and fatty acid biosynthesis exhibited increased activity in the OBE-ISR group after the treatment. Higher levels of acetate and propionate were found in the feces of the ISR group compared with the NOB and OBE groups. CONCLUSION The results of this study demonstrate that replacing rapidly digestible carbohydrates with slowly digestible carbohydrates within an HFD improve the composition of the gut microbiota. Consequently, metabolic disturbances associated with obesity may be prevented.
Collapse
Affiliation(s)
- Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Complejo Hospitalario Universitario de Granada, Granada, Spain
| | | | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Complejo Hospitalario Universitario de Granada, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Centre, University of Granada, Granada, Spain
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Munich, Germany
| | - Maria D. Giron
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | - Rafael Salto
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | | | | | | | | | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Complejo Hospitalario Universitario de Granada, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Centre, University of Granada, Granada, Spain
- CIBER Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Zhao Y, Zhong X, Yan J, Sun C, Zhao X, Wang X. Potential roles of gut microbes in biotransformation of natural products: An overview. Front Microbiol 2022; 13:956378. [PMID: 36246222 PMCID: PMC9560768 DOI: 10.3389/fmicb.2022.956378] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/29/2022] [Indexed: 11/23/2022] Open
Abstract
Natural products have been extensively applied in clinical practice, characterized by multi-component and multi-target, many pharmacodynamic substances, complex action mechanisms, and various physiological activities. For the oral administration of natural products, the gut microbiota and clinical efficacy are closely related, but this relationship remains unclear. Gut microbes play an important role in the transformation and utilization of natural products caused by the diversity of enzyme systems. Effective components such as flavonoids, alkaloids, lignans, and phenols cannot be metabolized directly through human digestive enzymes but can be transformed by enzymes produced by gut microorganisms and then utilized. Therefore, the focus is paid to the metabolism of natural products through the gut microbiota. In the present study, we systematically reviewed the studies about gut microbiota and their effect on the biotransformation of various components of natural products and highlighted the involved common bacteria, reaction types, pharmacological actions, and research methods. This study aims to provide theoretical support for the clinical application in the prevention and treatment of diseases and provide new ideas for studying natural products based on gut biotransformation.
Collapse
Affiliation(s)
- Yucui Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinqin Zhong
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junyuan Yan
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Congying Sun
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Zhao
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xin Zhao,
| | - Xiaoying Wang
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Xiaoying Wang,
| |
Collapse
|
19
|
Fermented Brewers’ Spent Grain Containing Dextran and Oligosaccharides as Ingredient for Composite Wheat Bread and Its Impact on Gut Metabolome In Vitro. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8100487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Brewers’ spent grain or BSG is a fiber and protein rich food-grade side stream that has remained underutilized due to its poor technological and sensory characteristics. In this study, BSG was fermented with Weissella confusa A16 in presence of sucrose to induce the synthesis of dextran and maltosyl-isomaltooligosaccharides. Fermented BSG with or without the above polysaccharides was used as ingredient in wheat bread. Digestion of BSG breads was simulated in vitro with Simulator of Human Intestinal Microbial Ecosystem, and levels of fecal metabolites were analyzed. Enrichment of BSG breads with in situ dextran and maltosyl-isomaltooligosaccharides improved the baking quality compared to native BSG. Metabolism of free amino acids and synthesis of short chain fatty acids varied at different stages and parts of colon. The increase in butyric acid was similar in both the proximal and distal colon. In situ dextran and maltosyl-isomaltooligosaccharides, and higher content of proteins and fiber in BSG breads had a positive influence towards gut microbiota functionality. Along with several essential amino acids, an increase in amount of γ-aminobutyric acid was also observed after simulated digestion. BSG breads had a significant effect on the gut metabolome during in vitro digestion, showing increased production of microbial metabolites with potential health benefits.
Collapse
|
20
|
Mary PR, Monica P, Kapoor M. Insights into β-manno-oligosaccharide uptake and metabolism in Bifidobacterium adolescentis DSMZ 20083 from whole-genome microarray analysis. Microbiol Res 2022; 266:127215. [DOI: 10.1016/j.micres.2022.127215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 10/14/2022]
|
21
|
Gadaleta RM, Cariello M, Crudele L, Moschetta A. Bile Salt Hydrolase-Competent Probiotics in the Management of IBD: Unlocking the "Bile Acid Code". Nutrients 2022; 14:3212. [PMID: 35956388 PMCID: PMC9370712 DOI: 10.3390/nu14153212] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/18/2023] Open
Abstract
Bile acid (BA) species and the gut microbiota (GM) contribute to intestinal mucosa homeostasis. BAs shape the GM and, conversely, intestinal bacteria with bile salt hydrolase (BSH) activity modulate the BA pool composition. The mutual interaction between BAs and intestinal microorganisms also influences mucosal barrier integrity, which is important for inflammatory bowel disease (IBD) pathogenesis, prevention and therapy. High levels of secondary BAs are detrimental for the intestinal barrier and increase the intestinal inflammatory response and dysbiosis. Additionally, a lack of BSH-active bacteria plays a role in intestinal inflammation and BA dysmetabolism. Thus, BSH-competent bacteria in probiotic formulations are being actively studied in IBD. At the same time, studies exploring the modulation of the master regulator of BA homeostasis, the Farnesoid X Receptor (FXR), in intestinal inflammation and how this impacts the GM are gaining significant momentum. Overall, the choice of probiotic supplementation should be a peculiar issue of personalized medicine, considering not only the disease but also the specific BA and metabolic signatures of a given patient.
Collapse
Affiliation(s)
- Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Lucilla Crudele
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
- INBB National Instituto for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|
22
|
Spore Powder of Paecilomyces hepiali Shapes Gut Microbiota to Relieve Exercise-Induced Fatigue in Mice. Nutrients 2022; 14:nu14142973. [PMID: 35889929 PMCID: PMC9323605 DOI: 10.3390/nu14142973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Paecilomyces hepiali, a fungal strain isolated from natural Ophiocordyceps sinensis, contains similar pharmacologically active components, has been used widely as a substitute of O. sinensis in functional food and medicine. However, the components and anti-fatigue effects of P.hepiali spores and their mechanisms of action are largely unknown. Here, we compared the chemical composition in P.hepiali spore (HPS) and mycelium (HPM) by liquid chromatography with tandem mass spectrometry analysis. We found 85 metabolites with significant differences, and HPS contains more L-Malic acid, Oxalacetic acid, Fructose-1,6-bisphosphate, and L-Arginine than HPM. Then we evaluated their anti-fatigue effects and regulatory effects on the gut microbiota in mice. The forced swimming time (SW) was only significantly increased in HPS groups: the high and low dose of the HPS group was 101% and 72% longer than the control group, respectively. Both HPS and HPM treatment decreased lactic acid, blood urea nitrogen, creatine kinase while increased lactate dehydrogenase (LDH) levels in the blood. Moreover, mice treated with HPS and HPM showed less skeletal muscle fiber spacing and breakage. The relative abundance of Alistips, Eubacterium, Bacterium, Parasutterella, and Olsenella in the gut microbiota of the HPS group was higher than that in the HPM group through 16S rRNA gene sequencing analysis. These changes may be related to the regulation of nucleotide, amino acid, and carbohydrate metabolism. Correlation analysis between the gut microbiota and fatigue-related indicators suggested that Alistips, Clostridium, Akkermansia, Olsenella, and Lactobacillus were positively correlated with the SW and LDH content. Our findings demonstrated that HPS has beneficial anti-fatigue effects by regulating gut microbiota.
Collapse
|
23
|
Effects of active, inactive, and derivatives of Akkermansia muciniphila on the expression of the endocannabinoid system and PPARs genes. Sci Rep 2022; 12:10031. [PMID: 35705595 PMCID: PMC9200819 DOI: 10.1038/s41598-022-13840-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/30/2022] [Indexed: 12/29/2022] Open
Abstract
This study aimed to investigate the effects of active and heat-inactivated forms of Akkermansia muciniphila, bacterium-derived outer membrane vesicles (OMVs), and cell-free supernatant on the transcription of endocannabinoid system (ECS) members, including cannabinoid receptors 1 and 2 (CB1 and CB2), fatty acid amide hydrolase (FAAH), and peroxisome proliferator-activated receptors (PPARs) genes (i.e., α, β/δ, and δ) in Caco-2 and HepG-2 cell lines. After the inoculation of A. muciniphila in brain heart infusion enriched medium, OMVs and cell-free supernatant were extracted. For the investigation of the effects of bacteria and its derivatives on the expression of ECS and PPARs genes, the aforementioned cells were treated by active and heat-inactivated bacteria, OMVs, and cell-free supernatant. Quantitative real-time polymerase chain reaction analysis revealed that both forms of the bacterium, bacterial-derived OMVs, and cell-free supernatant could affect the expression of CB1, CB2, FAAH, and PPARs genes (i.e., α, β/δ, and δ) significantly (P < 0.05). Considering the engagement of the aforementioned genes in metabolic pathways, it might be suggested that both forms of the bacterium, OMVs, and cell-free supernatant might have the potential to serve as a probiotic, paraprobiotic, and postbiotic candidate to prevent obesity, metabolic disorders, and liver diseases.
Collapse
|
24
|
Peng R, Han P, Fu J, Zhang ZW, Ma SR, Pan LB, Xia YY, Yu H, Xu H, Liu CX, Wang Y. Esterases From Bifidobacteria Exhibit the Conversion of Albiflorin in Gut Microbiota. Front Microbiol 2022; 13:880118. [PMID: 35464989 PMCID: PMC9019491 DOI: 10.3389/fmicb.2022.880118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/16/2022] [Indexed: 12/21/2022] Open
Abstract
Bifidobacteria is an important microbe that inhabits the human gut. It is capable of metabolizing complex compounds in the human diet. Albiflorin, an antidepressant natural product from Radix Paeoniae Alba in China, is difficult to absorb after oral administration, and its metabolism has been proven to be closely related to the gut microbiota. In this study, we demonstrated in vitro that several Bifidobacteria species were able to convert albiflorin to benzoic acid, and four esterases (B2, B3, B4, and BL) from Bifidobacterium breve and Bifidobacterium longum were found through genome mining and modeled by SWISS-MODEL. B2 and B3 presented the strongest albiflorin metabolism ability. The optimal conditions, including temperature, buffer, and pH, for the conversion of albiflorin by the four esterases were investigated. Furthermore, the effect of esterase on the metabolism of albiflorin in vivo was confirmed by transplanting bacteria containing esterase B2. This study demonstrated the vital role of esterases from Bifidobacteria in the metabolism of natural compounds containing ester bonds, which could contribute to the development of new enzymes, microbial evolution, and probiotic adjuvant compounds for treatment.
Collapse
Affiliation(s)
- Ran Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Fu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng-Wei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Rong Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Bin Pan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Yuan Xia
- Tianjin Institute of Pharmaceutical Research, Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Tianjin, China
| | - Hang Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang-Xiao Liu
- Tianjin Institute of Pharmaceutical Research, Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Tianjin, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Lee YM, Kim YJ. Two cases of bacteremic pneumonia caused by Bifidobacterium species. Anaerobe 2021; 73:102483. [PMID: 34839008 DOI: 10.1016/j.anaerobe.2021.102483] [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: 09/01/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 11/25/2022]
Abstract
Clinical infections by Bifidobacterium species rarely developed. We report two cases of bacteremic pneumonia caused by B. pseudocatenulatum and B. dentium, respectively, in patients vulnerable to aspiration. These cases suggested the potential for cause of serious pneumonia caused by Bifidobacterium species, in patients with high risk of aspiration.
Collapse
Affiliation(s)
- Yu-Mi Lee
- Division of Infectious Diseases, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Young Jin Kim
- Department of Laboratory Medicine, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
26
|
The Intestinal Dysbiosis of Mothers with Gestational Diabetes Mellitus (GDM) and Its Impact on the Gut Microbiota of Their Newborns. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2021; 2021:3044534. [PMID: 34603565 PMCID: PMC8481071 DOI: 10.1155/2021/3044534] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/11/2021] [Indexed: 02/06/2023]
Abstract
Gestational diabetes mellitus (GDM) is defined as “diagnosed as impaired glucose tolerance for the first time during pregnancy,” which can lead to adverse pregnancy outcomes and produces divergent effects on mothers and newborns. In recent years, with the continuous expansion of obese people, GDM shows an upward trend. The abundant and diverse members of the human gut microbiota exert critical roles in the maintenance of human health. Studies have shown that GDM may be associated with disordered gut microbiota in both mothers and newborns. Taking into account the potential effects on maternal and consequently neonatal health, in this review, we analyzed the available data and discussed the current knowledge about the potential relationship between GDM and intestinal dysbiosis in mothers and newborns. In addition, we also discussed the influencing factors derived from GDM mothers on the gut microbiome of their newborns, including the vertical transmission of microbiota from mothers, the alteration of milk components of GDM mothers, and using of probiotics. Hoping that new insights into the role of the gut microbiota in GDM could lead to the development of integrated strategies to prevent and treat these metabolic disorders.
Collapse
|
27
|
Cho HW, Eom YB. Forensic Analysis of Human Microbiome in Skin and Body Fluids Based on Geographic Location. Front Cell Infect Microbiol 2021; 11:695191. [PMID: 34458160 PMCID: PMC8388931 DOI: 10.3389/fcimb.2021.695191] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/26/2021] [Indexed: 01/16/2023] Open
Abstract
High-throughput DNA sequencing technologies have facilitated the in silico forensic analysis of human microbiome. Specific microbial species or communities obtained from the crime scene provide evidence of human contacts and their body fluids. The microbial community is influenced by geographic, ethnic, lifestyle, and environmental factors such as urbanization. An understanding of the effects of these external stressors on the human microbiome and determination of stable and changing elements are important in selecting appropriate targets for investigation. In this study, the Forensic Microbiome Database (FMD) (http://www.fmd.jcvi.org) containing the microbiome data of various locations in the human body in 35 countries was used. We focused on skin, saliva, vaginal fluid, and stool and found that the microbiome distribution differed according to the body part as well as the geographic location. In the case of skin samples, Staphylococcus species were higher than Corynebacterium species among Asians compared with Americans. Holdemanella and Fusobacterium were specific in the saliva of Koreans and Japanese populations. Lactobacillus was found in the vaginal fluids of individuals in all countries, whereas Serratia and Enterobacter were endemic to Bolivia and Congo, respectively. This study is the first attempt to collate and describe the observed variation in microbiomes from the forensic microbiome database. As additional microbiome databases are reported by studies worldwide, the diversity of the applications may exceed and expand beyond the initial identification of the host.
Collapse
Affiliation(s)
- Hye-Won Cho
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, South Korea
| | - Yong-Bin Eom
- Department of Medical Sciences, Graduate School, Soonchunhyang University, Asan, South Korea
- Department of Biomedical Laboratory Science, College of Medical Sciences, Soonchunhyang University, Asan, South Korea
| |
Collapse
|
28
|
The Mean of Milk: A Review of Human Milk Oligosaccharide Concentrations throughout Lactation. Nutrients 2021; 13:nu13082737. [PMID: 34444897 PMCID: PMC8398195 DOI: 10.3390/nu13082737] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
Human milk oligosaccharides (HMOs) are non-digestible and structurally diverse complex carbohydrates that are highly abundant in human milk. To date, more than 200 different HMO structures have been identified. Their concentrations in human milk vary according to various factors such as lactation period, mother’s genetic secretor status, and length of gestation (term or preterm). The objective of this review is to assess and rank HMO concentrations from healthy mothers throughout lactation at a global level. To this aim, published data from pooled (secretor and non-secretor) human milk samples were used. When samples were reported as secretor or non-secretor, means were converted to a pooled level, using the reported mean of approximately 80/20% secretor/non-secretor frequency in the global population. This approach provides an estimate of HMO concentrations in the milk of an average, healthy mother independent of secretor status. Mean concentrations of HMOs were extracted and categorized by pre-defined lactation periods of colostrum (0–5 days), transitional milk (6–14 days), mature milk (15–90 days), and late milk (>90 days). Further categorizations were made by gestational length at birth, mother’s ethnicity, and analytical methodology. Data were excluded if they were from preterm milk, unknown sample size and mothers with any known disease status. A total of 57 peer-reviewed articles reporting individual HMO concentrations published between 1996 and 2020 were included in the review. Pooled HMO means reported from 31 countries were analyzed. In addition to individual HMO concentrations, 12 articles reporting total HMO concentrations were also analyzed as a basis for relative HMO abundance. Total HMOs were found as 17.7 g/L in colostrum, 13.3 g/L in transitional milk, and 11.3 g/L in mature milk. The results show that HMO concentrations differ largely for each individual HMO and vary with lactation stages. For instance, while 2′-FL significantly decreased from colostrum (3.18 g/L ± 0.9) to late milk (1.64 g/L ± 0.67), 3-FL showed a significant increase from colostrum (0.37 g/L ± 0.1) to late milk (0.92 g/L ± 0.5). Although pooled human milk contains a diverse HMO profile with more than 200 structures identified, the top 10 individual HMOs make up over 70% of total HMO concentration. In mature pooled human milk, the top 15 HMOs in decreasing order of magnitude are 2′-FL, LNDFH-I (DFLNT), LNFP-I, LNFP-II, LNT, 3-FL, 6′-SL, DSLNT, LNnT, DFL (LDFT), FDS-LNH, LNFP-III, 3′-SL, LST c, and TF-LNH.
Collapse
|
29
|
Anker-Ladefoged C, Langkamp T, Mueller-Alcazar A. The Potential Impact of Selected Bacterial Strains on the Stress Response. Healthcare (Basel) 2021; 9:494. [PMID: 33922095 PMCID: PMC8143481 DOI: 10.3390/healthcare9050494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION The composition of the microbiome is subject to a variety of factors, such as eating behavior and the history of medical treatment. The interest in the impact of the microbiome on the stress response is mainly explained by the lack of development of new effective treatments for stress-related diseases. This scoping review aims to present the current state of research regarding the impact of bacterial strains in the gut on the stress response in humans in order to not only highlight these impacts but to also suggest potential intervention options. METHODS We included full-text articles on studies that: (a) were consistent with our research question; and (b) included the variable stress either using biomedical parameters such as cortisol or by examining the subjective stress level. Information from selected studies was synthesized from study designs and the main findings. RESULTS Seven studies were included, although they were heterogenous. The results of these studies do not allow a general statement about the effects of the selected bacterial strains on the stress response of the subjects and their precise pathways of action. However, one of the works gives evidence that the consumption of probiotics leads to a decrease in blood pressure and others show that stress-induced symptoms (including abdominal pain and headache) in healthy subjects could be reduced. CONCLUSION Due to different intake period and composition of the bacterial strains administered to the subjects, the studies presented here can only provide a limited meaningful judgement. As these studies included healthy participants between the ages of 18 and 60 years, a generalization to clinical populations is also not recommended. In order to confirm current effects and implement manipulation of the microbiome as a treatment method for clinical cases, future studies would benefit from examining the effects of the intestinal microbiome on the stress response in a clinical setting.
Collapse
Affiliation(s)
| | | | - Anett Mueller-Alcazar
- Department of Psychology, Faculty of Human Sciences, Medical School Hamburg, 20457 Hamburg, Germany; (C.A.-L.); (T.L.)
| |
Collapse
|
30
|
Phavichitr N, Wang S, Chomto S, Tantibhaedhyangkul R, Kakourou A, Intarakhao S, Jongpiputvanich S, Roeselers G, Knol J. Impact of synbiotics on gut microbiota during early life: a randomized, double-blind study. Sci Rep 2021; 11:3534. [PMID: 33574421 PMCID: PMC7878856 DOI: 10.1038/s41598-021-83009-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022] Open
Abstract
Human milk is considered the optimal nutrition for infants and found to contain significant numbers of viable bacteria. The aim of the study was to assess the effects of a specific synbiotic combination at doses closer to the bacterial cells present in human milk, on intestinal bifidobacteria proportions (relative abundance), reduction of potential pathogens and gut physiological conditions. A clinical study was conducted in 290 healthy infants aged from 6 to 19 weeks. Infants received either a control infant formula or one of the two investigational infant formulas (control formula with 0.8 g/100 ml scGOS/lcFOS and Bifidobacterium breve M-16V at either 1 × 104 cfu/ml or 1 × 106 cfu/ml). Exclusively breastfed infants were included as a reference. Analyses were performed on intention-to-treat groups and all-subjects-treated groups. After 6 weeks of intervention, the synbiotics at two different doses significantly increased the bifidobacteria proportions in healthy infants. The synbiotic supplementation also decreased the prevalence (infants with detectable levels) and the abundance of C. difficile. Closer to the levels in the breastfed reference group, fecal pH was significantly lower while l-lactate concentrations and acetate proportions were significantly higher in the synbiotic groups. All formulas were well tolerated and all groups showed a comparable safety profile based on the number and severity of adverse events and growth. In healthy infants, supplementation of infant-type bifidobacterial strain B. breve M-16V, at a dose close to bacterial numbers found in human milk, with scGOS/lcFOS (9:1) created a gut environment closer to the breastfed reference group. This specific synbiotic mixture may also support gut microbiota resilience during early life. Clinical Trial Registration This clinical study named Color Synbiotics Study, was registered in ClinicalTrials.gov on 18 March 2013. Registration number is NCT01813175. https://clinicaltrials.gov/ct2/show/NCT01813175.
Collapse
Affiliation(s)
| | - Shugui Wang
- Danone Nutricia Research, Singapore, Singapore
| | - Sirinuch Chomto
- Nutritional Unit, Department of Pediatrics, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Sukkrawan Intarakhao
- Department of Pediatrics, Thammasat Hospital, Faculty of Medicine, Thammasat University, Bangkok, Thailand
| | - Sungkom Jongpiputvanich
- Department of Pediatrics, Thammasat Hospital, Faculty of Medicine, Thammasat University, Bangkok, Thailand
| | | | | | - Jan Knol
- Danone Nutricia Research, Utrecht, The Netherlands.,Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
31
|
Gong C, Yang L, Liu K, Shen S, Zhang Q, Li H, Cheng Y. Effects of Antibiotic Treatment and Probiotics on the Gut Microbiome of 40 Infants Delivered Before Term by Cesarean Section Analysed by Using 16S rRNA Quantitative Polymerase Chain Reaction Sequencing. Med Sci Monit 2021; 27:e928467. [PMID: 33542172 PMCID: PMC7871509 DOI: 10.12659/msm.928467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/19/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effects on the gut microbiome of 40 infants delivered before term by cesarean section between antibiotic treatment and probiotics as assessed by 16S rRNA quantitative polymerase chain reaction (qPCR) sequencing. MATERIAL AND METHODS We divided 40 premature infants delivered by cesarean section into 4 groups according to exposure to antibiotics or probiotics: N group (No-probiotics and No-antibiotics), A group (antibiotics), P group (probiotics), and the AP group (antibiotics+probiotics). Fecal samples were collected on days 1, 3, and 10, and the microflora data were generated using 16S rRNA qPCR sequencing technology. The BugBase tool was used for phenotype prediction, the Tax4Fun tool was used for function prediction, and iPath software was used to predict the metabolic pathways of intestinal bacteria. RESULTS Antibiotics increased the abundance of pathogenic bacteria and reduced the replication and repair function (P=0.049), nucleotide metabolism function (P=0.047), and the purine metabolism pathways (P<0.05) of the gut microbiota. Probiotics increased the abundance of beneficial bacteria and the cellular community prokaryote function (P=0.042) and contributed to the Bifidobacteria biofilm formation. Probiotics alleviated the damage of antibiotics to the composition and function of the gut microbiota. CONCLUSIONS The findings from this study showed that antibiotic treatment of preterm infants born by cesarean section changed the gut microbiome, but that the use of probiotics could restore the normal microbiome, which supports that restoration of the normal gut microbiota may be achieved with probiotics.
Collapse
Affiliation(s)
- Chen Gong
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Liqi Yang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Kangkang Liu
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Shichun Shen
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Qixing Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Han Li
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yan Cheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
32
|
Barretto DA, Gadwala M, Vootla SK. The silkworm gut microbiota: A potential source for biotechnological applications. J Microbiol Methods 2021. [DOI: 10.1016/bs.mim.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
33
|
Alessandri G, Argentini C, Milani C, Turroni F, Cristina Ossiprandi M, van Sinderen D, Ventura M. Catching a glimpse of the bacterial gut community of companion animals: a canine and feline perspective. Microb Biotechnol 2020; 13:1708-1732. [PMID: 32864871 PMCID: PMC7533323 DOI: 10.1111/1751-7915.13656] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Dogs and cats have gained a special position in human society by becoming our principal companion animals. In this context, efforts to ensure their health and welfare have increased exponentially, with in recent times a growing interest in assessing the impact of the gut microbiota on canine and feline health. Recent technological advances have generated new tools to not only examine the intestinal microbial composition of dogs and cats, but also to scrutinize the genetic repertoire and associated metabolic functions of this microbial community. The application of high-throughput sequencing techniques to canine and feline faecal samples revealed similarities in their bacterial composition, with Fusobacteria, Firmicutes and Bacteroidetes as the most prevalent and abundant phyla, followed by Proteobacteria and Actinobacteria. Although key bacterial members were consistently present in their gut microbiota, the taxonomic composition and the metabolic repertoire of the intestinal microbial population may be influenced by several factors, including diet, age and anthropogenic aspects, as well as intestinal dysbiosis. The current review aims to provide a comprehensive overview of the multitude of factors which play a role in the modulation of the canine and feline gut microbiota and that of their human owners with whom they share the same environment.
Collapse
Affiliation(s)
- Giulia Alessandri
- Department of Veterinary Medical ScienceUniversity of ParmaParmaItaly
| | - Chiara Argentini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Maria Cristina Ossiprandi
- Department of Veterinary Medical ScienceUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| | - Douwe van Sinderen
- APC Microbiome Institute and School of Microbiology, Bioscience InstituteNational University of IrelandCorkIreland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental SustainabilityUniversity of ParmaParmaItaly
- Microbiome Research HubUniversity of ParmaParmaItaly
| |
Collapse
|
34
|
Gut Microbiota in Patients with Morbid Obesity Before and After Bariatric Surgery: a Ten-Year Review Study (2009-2019). Obes Surg 2020; 31:317-326. [PMID: 33130944 DOI: 10.1007/s11695-020-05074-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The changes in the composition and function of gut microbiota affect the metabolic functions (which are mediated by microbial effects) in patients with obesity, resulting in significant physiological regulation in these patients. Most of the studies emphasize that the Western-style diet (high fat and low vegetable consumption) leads to significant changes in the intestinal microbiome in individuals with metabolic syndrome. A deeper understanding of the profiles of gut microbes will contribute to the development of new therapeutic strategies for the management of metabolic syndrome and other metabolic diseases and related disorders. The aim of this review is to evaluate recent experimental evidence outlining the alterations of gut microbiota composition and function in recovery from bariatric surgical operations with an emphasis on sleeve gastrectomy and gastric bypass.
Collapse
|
35
|
Hori T, Matsuda K, Oishi K. Probiotics: A Dietary Factor to Modulate the Gut Microbiome, Host Immune System, and Gut-Brain Interaction. Microorganisms 2020; 8:microorganisms8091401. [PMID: 32933067 PMCID: PMC7563712 DOI: 10.3390/microorganisms8091401] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/26/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Various benefits of probiotics to the host have been shown in numerous human clinical trials. These organisms have been proposed to act by improving the balance of the gut microbiota and enhancing the production of short-chain fatty acids, as well as by interacting with host cells in the gastrointestinal tract, including immune cells, nerve cells, and endocrine cells. Although the stimulation of host cells by probiotics and subsequent signaling have been explained by in vitro experiments and animal studies, there has been some skepticism as to whether probiotics can actually interact with host cells in the human gastrointestinal tract, where miscellaneous indigenous bacteria coexist. Most recently, it has been shown that the ileal microbiota in humans after consumption of a fermented milk is occupied by probiotics for several hours, indicating that there is adequate opportunity for the ingested strain to stimulate the host cells continuously over a period of time. As the dynamics of ingested probiotics in the human gastrointestinal tract become clearer, further progress in this research area is expected to elucidate their behavior within the tract, as well as the mechanism of their physiological effects on the host.
Collapse
|
36
|
Draft Genome Sequences of Two Strains of Bifidobacterium dentium Isolated from a Crude Fecal Extract Used for Fecal Microbiota Transplantation in the Republic of Korea. Microbiol Resour Announc 2020; 9:9/23/e00197-20. [PMID: 32499362 PMCID: PMC7272543 DOI: 10.1128/mra.00197-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We present the draft genome sequences of two Bifidobacterium dentium strains isolated from a fecal extract for fecal microbiota transplantation at a hospital in the Republic of Korea. Phylogenetic and functional analyses were performed to understand the physiological characteristics and functions of Bifidobacterium spp. in the human intestine. We present the draft genome sequences of two Bifidobacterium dentium strains isolated from a fecal extract for fecal microbiota transplantation at a hospital in the Republic of Korea. Phylogenetic and functional analyses were performed to understand the physiological characteristics and functions of Bifidobacterium spp. in the human intestine.
Collapse
|
37
|
Abstract
The gastrointestinal microbiome plays a pivotal role in physiological homeostasis of the intestine as well as in the pathophysiology of diseases including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Emerging evidence suggests that gut microbiota signal to the mitochondria of mucosal cells, including epithelial cells and immune cells. Gut microbiota signaling to mitochondria has been shown to alter mitochondrial metabolism, activate immune cells, induce inflammasome signaling, and alter epithelial barrier function. Both dysbiosis of the gut microbiota and mitochondrial dysfunction are associated with chronic intestinal inflammation and CRC. This review discusses mitochondrial metabolism of gut mucosal cells, mitochondrial dysfunction, and known gut microbiota-mediated mitochondrial alterations during IBD and CRC.
Collapse
Affiliation(s)
- Dakota N. Jackson
- Department of Internal Medicine, Division of Gastroenterology, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Arianne L. Theiss
- Department of Internal Medicine, Division of Gastroenterology, Baylor Scott & White Research Institute, Baylor University Medical Center, Dallas, TX, USA,CONTACT Arianne L. Theiss Division of Gastroenterology, Baylor Research Institute, Baylor University Medical Center, 250 Hoblitzelle, 3500 Gaston Avenue, Dallas, TX75246, USA
| |
Collapse
|
38
|
Wong CB, Odamaki T, Xiao JZ. Insights into the reason of Human-Residential Bifidobacteria (HRB) being the natural inhabitants of the human gut and their potential health-promoting benefits. FEMS Microbiol Rev 2020; 44:369-385. [PMID: 32319522 PMCID: PMC7326374 DOI: 10.1093/femsre/fuaa010] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
Members of Bifidobacterium are among the first microbes to colonise the human gut, and certain species are recognised as the natural resident of human gut microbiota. Their presence in the human gut has been associated with health-promoting benefits and reduced abundance of this genus is linked with several diseases. Bifidobacterial species are assumed to have coevolved with their hosts and include members that are naturally present in the human gut, thus recognised as Human-Residential Bifidobacteria (HRB). The physiological functions of these bacteria and the reasons why they occur in and how they adapt to the human gut are of immense significance. In this review, we provide an overview of the biology of bifidobacteria as members of the human gut microbiota and address factors that contribute to the preponderance of HRB in the human gut. We highlight some of the important genetic attributes and core physiological traits of these bacteria that may explain their adaptive advantages, ecological fitness, and competitiveness in the human gut. This review will help to widen our understanding of one of the most important human commensal bacteria and shed light on the practical consideration for selecting bifidobacterial strains as human probiotics.
Collapse
Affiliation(s)
- Chyn Boon Wong
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa, 252–8583 Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa, 252–8583 Japan
| | - Jin-zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., 5-1-83, Higashihara, Zama, Kanagawa, 252–8583 Japan
| |
Collapse
|
39
|
Kapourchali FR, Cresci GAM. Early-Life Gut Microbiome-The Importance of Maternal and Infant Factors in Its Establishment. Nutr Clin Pract 2020; 35:386-405. [PMID: 32329544 DOI: 10.1002/ncp.10490] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
The early-life microbiome is gaining appreciation as a major influencer in human development and long-term health. Multiple factors are known to influence the initial colonization, development, and function of the neonatal gut microbiome. In addition, alterations in early-life gut microbial composition is associated with several chronic health conditions such as obesity, asthma, and allergies. In this review, we focus on both maternal and infant factors known to influence early-life gut colonization. Also reviewed is the important role of infant feeding, including evidence-based strategies for maternal and infant supplementation with the goal to protect and/or restore the infant gut microbiome.
Collapse
Affiliation(s)
| | - Gail A M Cresci
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Human Nutrition, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
40
|
Turroni F, Milani C, Duranti S, Lugli GA, Bernasconi S, Margolles A, Di Pierro F, van Sinderen D, Ventura M. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr 2020; 46:16. [PMID: 32024556 PMCID: PMC7003403 DOI: 10.1186/s13052-020-0781-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022] Open
Abstract
Initial establishment of the human gut microbiota is generally believed to occur immediately following birth, involving key gut commensals such as bifidobacteria that are acquired from the mother. The subsequent development of this early gut microbiota is driven and modulated by specific dietary compounds present in human milk that support selective colonization. This represents a very intriguing example of host-microbe co-evolution, where both partners are believed to benefit. In recent years, various publications have focused on dissecting microbial infant gut communities and their interaction with their human host, being a determining factor in host physiology and metabolic activities. Such studies have highlighted a reduction of microbial diversity and/or an aberrant microbiota composition, sometimes referred to as dysbiosis, which may manifest itself during the early stage of life, i.e., in infants, or later stages of life. There are growing experimental data that may explain how the early human gut microbiota affects risk factors related to adult health conditions. This concept has fueled the development of various nutritional strategies, many of which are based on probiotics and/or prebiotics, to shape the infant microbiota. In this review, we will present the current state of the art regarding the infant gut microbiota and the role of key commensal microorganisms like bifidobacteria in the establishment of the first microbial communities in the human gut.
Collapse
Affiliation(s)
- Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | - Gabriele Andrea Lugli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy
| | | | - Abelardo Margolles
- Departamento de Microbiologia y Bioquimica de Productos Lacteos, IPLA - CSIC, Villaviciosa, Asturias, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias-ISPA, Oviedo, Spain
| | | | - Douwe van Sinderen
- School of Microbiology & APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11a, 43124, Parma, Italy.
- Microbiome Research Hub, University of Parma, Parma, Italy.
| |
Collapse
|
41
|
Pourteymour Fard Tabrizi F, Abbasalizad Farhangi M. A Systematic Review of the Potential Effects of Thylakoids in the Management of Obesity and Its Related Issues. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2019.1710747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Fatemeh Pourteymour Fard Tabrizi
- Student Research Committee, Department of Nutrition, Faculty of Nutrition and Food science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Abbasalizad Farhangi
- Research Center for Evidence Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
El Hadad S, Zakareya A, Al-Hejin A, Aldahlawi A, Alharbi M. Sustaining exposure to high concentrations of bifidobacteria inhibits gene expression of Mouse's mucosal immunity. Heliyon 2019; 5:e02866. [PMID: 31890933 PMCID: PMC6926234 DOI: 10.1016/j.heliyon.2019.e02866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/23/2019] [Accepted: 11/12/2019] [Indexed: 12/28/2022] Open
Abstract
Numerous dietary products are supplemented with probiotics that may be beneficial for human health. Recently, bifidobacteria have received increasing attention as a genus of probiotic bacteria with high efficiency and few side effects. To examine potential effects of different bifidobacteria concentrations on the mucosal immune response, we fed mice with (a) 108 colony-forming units (CFU) of bifidobacteria (group 108B), and (b) with 1012 CFU of bifidobacteria (group 1012B) over 42 days and assessed gene expression in intestinal mucosa and immune marker concentrations in serum samples; ten untreated female mice were used as a control. Continuous exposure to 108 CFU of bifidobacteria activated both macrophages and Treg immune cells through significantly increasing the expression of mucosal TLR2 and IL10-mRNA genes, but inhibited Th1 and Th2 cells via significant downregulation of IL4 and IFNγ gene expression, compared to untreated mice. Interestingly, group 1012B showed down-regulated expression of TLR2, IL10, and IL4 genes but up-regulated expression of IFNγ, compared to group 108B and to the control. Also, polyclonal immunoglobulins IgG, IgM, and IgA showed a significant increase in all treated mice compared to the control. We conclude that high concentrations of bifidobacteria reduced innate immune functions. Furthermore, adaptive immunity seemed to be enhanced by increasing stimulation of T and B lymphocytes, suggesting aberration of the immune system following intestinal inflammation due to constant exposure to high concentrations of bifidobacteria. Both experimental bifidobacteria concentrations increased the total levels of circulating Igs, particularly of IgA.
Collapse
Affiliation(s)
- Sahar El Hadad
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Research Center of Genetic Engineering and Bioinformatics, VACSERA, Cairo, Egypt.,Immunolgy Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayeshah Zakareya
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Al-Hejin
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Microbiology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alia Aldahlawi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Immunolgy Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Alharbi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
43
|
Baquero F, Lanza VF, Baquero MR, Del Campo R, Bravo-Vázquez DA. Microcins in Enterobacteriaceae: Peptide Antimicrobials in the Eco-Active Intestinal Chemosphere. Front Microbiol 2019; 10:2261. [PMID: 31649628 PMCID: PMC6795089 DOI: 10.3389/fmicb.2019.02261] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Microcins are low-molecular-weight, ribosomally produced, highly stable, bacterial-inhibitory molecules involved in competitive, and amensalistic interactions between Enterobacteriaceae in the intestine. These interactions take place in a highly complex chemical landscape, the intestinal eco-active chemosphere, composed of chemical substances that positively or negatively influence bacterial growth, including those originated from nutrient uptake, and those produced by the action of the human or animal host and the intestinal microbiome. The contribution of bacteria results from their effect on the host generated molecules, on food and digested food, and organic substances from microbial origin, including from bacterial degradation. Here, we comprehensively review the main chemical substances present in the human intestinal chemosphere, particularly of those having inhibitory effects on microorganisms. With this background, and focusing on Enterobacteriaceae, the most relevant human pathogens from the intestinal microbiota, the microcin’s history and classification, mechanisms of action, and mechanisms involved in microcin’s immunity (in microcin producers) and resistance (non-producers) are reviewed. Products from the chemosphere likely modulate the ecological effects of microcin activity. Several cross-resistance mechanisms are shared by microcins, colicins, bacteriophages, and some conventional antibiotics, which are expected to produce cross-effects. Double-microcin-producing strains (such as microcins MccM and MccH47) have been successfully used for decades in the control of pathogenic gut organisms. Microcins are associated with successful gut colonization, facilitating translocation and invasion, leading to bacteremia, and urinary tract infections. In fact, Escherichia coli strains from the more invasive phylogroups (e.g., B2) are frequently microcinogenic. A publicly accessible APD3 database http://aps.unmc.edu/AP/ shows particular genes encoding microcins in 34.1% of E. coli strains (mostly MccV, MccM, MccH47, and MccI47), and much less in Shigella and Salmonella (<2%). Some 4.65% of Klebsiella pneumoniae are microcinogenic (mostly with MccE492), and even less in Enterobacter or Citrobacter (mostly MccS). The high frequency and variety of microcins in some Enterobacteriaceae indicate key ecological functions, a notion supported by their dominance in the intestinal microbiota of biosynthetic gene clusters involved in the synthesis of post-translationally modified peptide microcins.
Collapse
Affiliation(s)
- Fernando Baquero
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Val F Lanza
- Bioinformatics Unit, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Maria-Rosario Baquero
- Department of Microbiology, Alfonso X El Sabio University, Villanueva de la Cañada, Spain
| | - Rosa Del Campo
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - Daniel A Bravo-Vázquez
- Department of Microbiology, Alfonso X El Sabio University, Villanueva de la Cañada, Spain
| |
Collapse
|
44
|
Rett Syndrome and Other Neurodevelopmental Disorders Share Common Changes in Gut Microbial Community: A Descriptive Review. Int J Mol Sci 2019; 20:ijms20174160. [PMID: 31454888 PMCID: PMC6747313 DOI: 10.3390/ijms20174160] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
In this narrative review, we summarize recent pieces of evidence of the role of microbiota alterations in Rett syndrome (RTT). Neurological problems are prominent features of the syndrome, but the pathogenic mechanisms modulating its severity are still poorly understood. Gut microbiota was recently demonstrated to be altered both in animal models and humans with different neurodevelopmental disorders and/or epilepsy. By investigating gut microbiota in RTT cohorts, a less rich microbial community was identified which was associated with alterations of fecal microbial short-chain fatty acids. These changes were positively correlated with severe clinical outcomes. Indeed, microbial metabolites can play a crucial role both locally and systemically, having dynamic effects on host metabolism and gene expression in many organs. Similar alterations were found in patients with autism and down syndrome as well, suggesting a potential common pathway of gut microbiota involvement in neurodevelopmental disorders.
Collapse
|
45
|
Genome Sequence and Biochemical Properties of Bifidobacterium longum Strain ICIS-505, Isolated from the Intestine of a Healthy Woman. Microbiol Resour Announc 2019; 8:8/33/e00491-19. [PMID: 31416861 PMCID: PMC6696636 DOI: 10.1128/mra.00491-19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This report describes the genome sequence of Bifidobacterium longum strain ICIS-505, isolated from human feces. The size of the genome was 2,448,844 bp (59.71% G+C content), including 3,751 bp of the crypto-plasmid pBL505. Annotation revealed 2,241 gene sequences, including 2,033 proteins, 7 rRNA genes, 76 tRNA genes, and 4 noncoding RNA genes. This report describes the genome sequence of Bifidobacterium longum strain ICIS-505, isolated from human feces. The size of the genome was 2,448,844 bp (59.71% G+C content), including 3,751 bp of the crypto-plasmid pBL505. Annotation revealed 2,241 gene sequences, including 2,033 proteins, 7 rRNA genes, 76 tRNA genes, and 4 noncoding RNA genes.
Collapse
|
46
|
Hao H, Zhu L, Faden HS. The milk-based diet of infancy and the gut microbiome. Gastroenterol Rep (Oxf) 2019; 7:246-249. [PMID: 31413830 PMCID: PMC6688733 DOI: 10.1093/gastro/goz031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/05/2019] [Accepted: 07/04/2019] [Indexed: 01/07/2023] Open
Abstract
The composition and the diversity of the gut microbiome play a major role in the health and well-being of humans beginning at birth. The impact of the diet on the structure and the function of the gut microbiome is evident by the changes in the gut microbiome concurrent with the transition from human milk to solid food. Complex oligosaccharides contained in milk are essential nutrients for commensal microbes in the infant gut. The most important commensal bacterium in the infant gut, bifidobacterium, requires α1, 2 fucosylated oligosaccharides for growth. Because not all humans are able to secrete α1, 2 fucosylated oligosaccharides into milk, the gut microbiome of infants and bifidobacteria, in particular, vary considerably between ‘secretors’ and ‘non-secretors’. A paucity of α1, 2 fucosylated oligosaccharides and bifidobacteria in the gut of infants may be associated with poor health.
Collapse
Affiliation(s)
- Hu Hao
- Division of Pediatrics, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Lixin Zhu
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Biochemistry, Genome, Environment and Microbiome Community of Excellence, State University of New York at Buffalo, Buffalo, NY, USA
| | - Howard S Faden
- Division of Infectious Diseases, Department of Pediatrics, Jacobs School of Medicine and Biological Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Celiac disease (CD) is an autoimmune enteropathy triggered by gluten. The purpose of this review is to examine the major genetic and environmental factors that contribute to CD pathogenesis. RECENT FINDINGS We reviewed the current state of knowledge on the genetic and environmental components that play a role in CD onset. A genome-wide association study (GWAS) analysis has highlighted several genes other than HLA involved in CD. Recent studies have shown that HLA haplotype influences the microbiome composition in infants and that dysbiosis in the intestinal microflora, in turn, contributes to loss of tolerance to gluten. Recently, observational studies have discussed the hypothesis stating that breast-feeding had a protective role against CD onset. CD etiology is influenced by genetic and environmental factors. A better understanding of these components would deepen our knowledge on the mechanisms that lead to loss of tolerance and could help in developing a more "personalized medicine."
Collapse
|
48
|
Coker MO, Hoen AG, Dade E, Lundgren S, Li Z, Wong AD, Zens MS, Palys TJ, Morrison HG, Sogin ML, Baker ER, Karagas MR, Madan JC. Specific class of intrapartum antibiotics relates to maturation of the infant gut microbiota: a prospective cohort study. BJOG 2019; 127:217-227. [PMID: 31006170 DOI: 10.1111/1471-0528.15799] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To evaluate the potential impact of intrapartum antibiotics, and their specific classes, on the infant gut microbiota in the first year of life. DESIGN Prospective study of infants in the New Hampshire Birth Cohort Study (NHBCS). SETTINGS Rural New Hampshire, USA. POPULATION OR SAMPLE Two hundred and sixty-six full-term infants from the NHBCS. METHODS Intrapartum antibiotic use during labour and delivery was abstracted from medical records. Faecal samples collected at 6 weeks and 1 year of age were characterised by 16S rRNA sequencing, and metagenomics analysis in a subset of samples. EXPOSURES Maternal exposure to antibiotics during labour and delivery. MAIN OUTCOME MEASURE Taxonomic and functional profiles of faecal samples. RESULTS Infant exposure to intrapartum antibiotics, particularly to two or more antibiotic classes, was independently associated with lower microbial diversity scores as well as a unique bacterial community at 6 weeks (GUnifrac, P = 0.02). At 1 year, infants in the penicillin-only group had significantly lower α diversity scores than infants not exposed to intrapartum antibiotics. Within the first year of life, intrapartum exposure to penicillins was related to a significantly lower increase in several taxa including Bacteroides, use of cephalosporins was associated with a significantly lower rise over time in Bifidobacterium and infants in the multi-class group experienced a significantly higher increase in Veillonella dispar. CONCLUSIONS Our findings suggest that intrapartum antibiotics alter the developmental trajectory of the infant gut microbiome, and specific antibiotic types may impact community composition, diversity and keystone immune training taxa. TWEETABLE ABSTRACT Class of intrapartum antibiotics administered during delivery relates to maturation of infant gut microbiota.
Collapse
Affiliation(s)
- M O Coker
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - A G Hoen
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - E Dade
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - S Lundgren
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Z Li
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - A D Wong
- The Dartmouth Institute, Lebanon, NH, USA
| | - M S Zens
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - T J Palys
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - H G Morrison
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
| | - M L Sogin
- Josephine Bay Paul Center, Marine Biological Laboratory, Woods Hole, MA, USA
| | - E R Baker
- Division of Neonatology, Department of Pediatrics, Children's Hospital at Dartmouth, Lebanon, NH, USA
| | - M R Karagas
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Center for Molecular Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - J C Madan
- Department of Epidemiology, The Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Division of Neonatology, Department of Pediatrics, Children's Hospital at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
49
|
Li Z, Wang G, Zhang K, Gong W, Yu E, Tian J, Xie J, Yu D. Epizootic ulcerative syndrome causes cutaneous dysbacteriosis in hybrid snakehead ( Channa maculata♀ × Channa argus♂). PeerJ 2019; 7:e6674. [PMID: 30972254 PMCID: PMC6450373 DOI: 10.7717/peerj.6674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/23/2019] [Indexed: 01/09/2023] Open
Abstract
Cutaneous microbiota play an important role in protecting fish against pathogens. Aphanomyces infection causes epizootic ulcerative syndrome (EUS) in fish, and by perturbing the integrity of the cutaneous microbiota, increases the potential for infection by pathogenic bacteria. However, whether the composition of the cutaneous microbiota is altered in fish with EUS, and if so, which species are changed and how this might influence infected fish, is still largely unclear. Considering the importance of cutaneous microbiota in maintaining host health, we hypothesized that Aphanomyces infection significantly enhances the presence of certain bacterial pathogens in the cutaneous microbiota and causes cutaneous dysbacteriosis. To test this hypothesis, we compared the cutaneous microbiota compositions of hybrid snakehead (Channa maculata♀ × Channa argus♂) with and without Aphanomyces infection using Illumina Miseq sequencing of the 16S rRNA gene. Our results showed that the cutaneous microbiota of hybrid snakehead were significantly altered subsequent to EUS infection and that the numbers of potentially pathogenic bacteria classified into the genera Anaerosinus, Anaerovorax, Dorea, and Clostridium were significantly enhanced in the cutaneous microbiota of hybrid snakehead with EUS, whereas bacteria classified into the genera Arthrobacter, Dysgonomonas, Anoxybacillus, Bacillus, Solibacillus, Carnobacterium, Lactococcus, Streptococcus, Achromobacter, Polynucleobacter, Vogesella, and Pseudomonas were significantly reduced. These results imply that treatment for EUS should not only take into consideration the control of Aphanomyces reproduction but should also focus on regulating the cutaneous microbiota of infected fish.
Collapse
Affiliation(s)
- Zhifei Li
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Guangjun Wang
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Kai Zhang
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Wangbao Gong
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Ermeng Yu
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Jingjing Tian
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Jun Xie
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| | - Deguang Yu
- Key Laboratory of Tropical and Subtropical Fishery Resource Application and Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China.,Guangdong Ecological Remediation of Aquaculture Pollution Research Center, Guangzhou, China
| |
Collapse
|
50
|
Jazani NH, Savoj J, Lustgarten M, Lau WL, Vaziri ND. Impact of Gut Dysbiosis on Neurohormonal Pathways in Chronic Kidney Disease. Diseases 2019; 7:diseases7010021. [PMID: 30781823 PMCID: PMC6473882 DOI: 10.3390/diseases7010021] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/29/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a worldwide major health problem. Traditional risk factors for CKD are hypertension, obesity, and diabetes mellitus. Recent studies have identified gut dysbiosis as a novel risk factor for the progression CKD and its complications. Dysbiosis can worsen systemic inflammation, which plays an important role in the progression of CKD and its complications such as cardiovascular diseases. In this review, we discuss the beneficial effects of the normal gut microbiota, and then elaborate on how alterations in the biochemical environment of the gastrointestinal tract in CKD can affect gut microbiota. External factors such as dietary restrictions, medications, and dialysis further promote dysbiosis. We discuss the impact of an altered gut microbiota on neuroendocrine pathways such as the hypothalamus⁻pituitary⁻adrenal axis, the production of neurotransmitters and neuroactive compounds, tryptophan metabolism, and the cholinergic anti-inflammatory pathway. Finally, therapeutic strategies including diet modification, intestinal alpha-glucosidase inhibitors, prebiotics, probiotics and synbiotics are reviewed.
Collapse
Affiliation(s)
- Nima H Jazani
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| | - Javad Savoj
- Department of Internal Medicine, Riverside Community Hospital, University of California-Riverside School of Medicine, Riverside, CA 92501, USA.
| | - Michael Lustgarten
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| | - Wei Ling Lau
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| | - Nosratola D Vaziri
- Division of Nephrology, Department of Medicine, University of California-Irvine, Irvine, CA 92697, USA.
| |
Collapse
|